51
|
Tsai CL, Erickson KI, Sun HS, Kuo YM, Pai MC. A cross-sectional examination of a family history of Alzheimer's disease and ApoE epsilon 4 on physical fitness, molecular biomarkers, and neurocognitive performance. Physiol Behav 2020; 230:113268. [PMID: 33383402 DOI: 10.1016/j.physbeh.2020.113268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/30/2020] [Accepted: 11/21/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE The present study examined whether the ɛ4 allele of the apolipoprotein E (ApoE) gene impacts molecular biomarkers and neurocognitive performance among individuals at genetic risk for developing Alzheimer's disease (AD). The correlations between physical fitness and molecular/neurocognitive indices were also explored. METHODS Fasting blood samples were collected from 162 individuals with a family history of AD (ADFH). There were twenty-two carriers of the ApoE-4 variant (ApoE-4 group). For comparison purposes we randomly selected 22 non-ɛ4 carriers (non-ApoE-4 group) from the ADFH individuals. Circulating inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6, IL-8, and IL-15), neuroprotective growth factors (e.g., BDNF, IGF-1, IGF-2, VEGF, and FGF-2), and Amyloid-β peptides (e.g., Aβ1-40 and Aβ1-42), neurocognitive performance [e.g., behavior and brain even-related potentials (ERP)] during a task-switching paradigm, as well as physical fitness scores were measured. RESULTS The ApoE-4 group relative to the non-ApoE-4 group was similar with respect to molecular biomarkers, physical fitness, and most measures of neurocognitive performance. However, ADFH individuals that were ɛ4 carriers exhibited significantly higher local switching accuracy costs, worse accuracy as well as smaller ERP P3 amplitudes for the memory-switching condition. Importantly, cardiorespiratory fitness levels were significantly correlated with accuracy for most task-switching conditions, and levels of BDNF, Aβ1-40, and Aβ1-42 collapsed across the two groups even when controlling for the age co-variable, while the ApoE-4 group revealed similar pattern of results. CONCLUSIONS These data suggest that individuals with ADFH that were carriers of the ApoE-4 variant performed worse on the task-switching paradigm and that this could be due to compromised task-set and memory updating processes. Physical exercise interventions aimed to enhance cardiorespiratory fitness levels could be a potential AD prevention strategy for ameliorating cognitive function and reducing the accumulation of the Aβ peptides in this high risk group.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Taiwan.
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, USA; Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Western Australia
| | - H-Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Taiwan.
| |
Collapse
|
52
|
Does APOE genotype moderate the relationship between physical activity, brain health and dementia risk? A systematic review. Ageing Res Rev 2020; 64:101173. [PMID: 32961338 DOI: 10.1016/j.arr.2020.101173] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/09/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION For decades, researchers have tried to understand the moderating effect of APOE ε4 carriage on the relationship between physical activity (PA), brain health and dementia risk. However, this field has produced inconsistent findings. METHOD We conducted a systematic review of the literature, searching for observational and interventional studies examining the effect of APOE ε4 carriage on the relationships between PA, dementia risk and different markers of brain health. RESULTS Observational studies using dementia risk as a primary outcome measure generally found that in shorter follow-up periods (up to 10 years) both APOE ε4 carriers and non-carriers benefit from PA, although longer follow-ups showed mixed results. In neuroimaging studies, mainly carriers or both groups showed benefits. Additionally, the association between PA and amyloid burden was more evident among carriers. Overall, studies with greater samples of active APOE ε4 carriers are more likely to report benefits within this group in terms of lower dementia risk and reduced brain pathology. DISCUSSION Although we have identified some patterns for the modulating effect of APOE ε4 on PA and dementia or brain pathology, the available data is, overall, inconclusive. Heterogeneity in study design, methodology, and outcomes blur the ability to detect clear associations.
Collapse
|
53
|
Tarumi T, Rossetti H, Thomas BP, Harris T, Tseng BY, Turner M, Wang C, German Z, Martin-Cook K, Stowe AM, Womack KB, Mathews D, Kerwin DR, Hynan L, Diaz-Arrastia R, Lu H, Cullum CM, Zhang R. Exercise Training in Amnestic Mild Cognitive Impairment: A One-Year Randomized Controlled Trial. J Alzheimers Dis 2020; 71:421-433. [PMID: 31403944 DOI: 10.3233/jad-181175] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The current evidence is inconclusive to support the benefits of aerobic exercise training (AET) for preventing neurocognitive decline in patients with amnestic mild cognitive impairment (aMCI). OBJECTIVE To examine the effect of a progressive, moderate-to-high intensity AET program on memory and executive function, brain volume, and cortical amyloid-β (Aβ) plaque deposition in aMCI patients. METHODS This is a proof-of-concept trial that randomized 70 aMCI patients to 12 months of AET or stretching and toning (SAT, active control) interventions. Primary neuropsychological outcomes were assessed by using the California Verbal Learning Test-second edition (CVLT-II) and the Delis-Kaplan Executive Function System (D-KEFS). Secondary outcomes were the global and hippocampal brain volumes and the mean cortical and precuneus Aβ deposition. RESULTS Baseline cognitive scores were similar between the groups. Memory and executive function performance improved over time but did not differ between the AET and SAT groups. Brain volume decreased and precuneus Aβ plaque deposition increased over time but did not differ between the groups. Cardiorespiratory fitness was significantly improved in the AET compared with SAT group. In amyloid positive patients, AET was associated with reduced hippocampal atrophy when compared with the SAT group. CONCLUSION The AET and SAT groups both showed evidence of slightly improved neuropsychological scores in previously sedentary aMCI patients. However, these interventions did not prevent brain atrophy or increases in cortical Aβ deposition over 12 months. In amyloid positive patients, AET reduced hippocampal atrophy when compared with the SAT group.
Collapse
Affiliation(s)
- Takashi Tarumi
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA.,Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Human Informatics Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Heidi Rossetti
- Departments of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Binu P Thomas
- Departments of Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas Harris
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin Y Tseng
- Department of Health and Kinesiology, The University of Texas at Tyler, Tyler, TX, USA
| | - Marcel Turner
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA
| | - Ciwen Wang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA
| | - Zohre German
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kristin Martin-Cook
- Departments of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ann M Stowe
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kyle B Womack
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dana Mathews
- Departments of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana R Kerwin
- Institute for Texas Alzheimer's and Memory Disorders, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA
| | - Linda Hynan
- Departments of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ramon Diaz-Arrastia
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hanzhang Lu
- Departments of Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - C Munro Cullum
- Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA.,Departments of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
54
|
Fraser MA, Walsh EI, Shaw ME, Abhayaratna WP, Anstey KJ, Sachdev PS, Cherbuin N. Longitudinal trajectories of hippocampal volume in middle to older age community dwelling individuals. Neurobiol Aging 2020; 97:97-105. [PMID: 33190123 DOI: 10.1016/j.neurobiolaging.2020.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/04/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Understanding heterogeneity in brain aging trajectories is important to estimate the extent to which aging outcomes can be optimized. Although brain changes in late life are well-characterized, brain changes in middle age are not well understood. In this study, we investigated hippocampal change in a generally healthy community-living population of middle (n = 421, mean age 47.2 years) and older age (n = 411, mean age 63.0 years) individuals, over a follow-up of up to 12 years. Manually traced hippocampal volumes were analyzed using multilevel models and latent class analysis to investigate longitudinal aging trajectories and laterality and sex effects, and to identify subgroups that follow different aging trajectories. Hippocampal volumes decreased on average by 0.18%/year in middle age and 0.3%/year in older age. Men tended to experience steeper declines than women in middle age only. Three subgroups of individuals following different trajectories were identified in middle age and 2 in older age. Contrary to expectations, the subgroup containing two-thirds of older age participants maintained stable hippocampal volumes across the follow-up.
Collapse
Affiliation(s)
- Mark A Fraser
- Centre for Research on Ageing, Health and Wellbeing, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia.
| | - Erin I Walsh
- Centre for Research on Ageing, Health and Wellbeing, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia; Population Health Exchange, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Marnie E Shaw
- ANU College of Engineering & Computer Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Walter P Abhayaratna
- College of Health & Medicine, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kaarin J Anstey
- Centre for Research on Ageing, Health and Wellbeing, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia; Ageing Futures Institute, University of New South Wales, Sydney, New South Wales, Australia; Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, Research School of Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
55
|
Palta P, Rippon B, Reitz C, He H, Sherwood G, Ceballos F, Teresi J, Razlighi Q, Moreno H, Brickman AM, Luchsinger JA. Apolipoprotein E genotype and in vivo amyloid burden in middle-aged Hispanics. Neurology 2020; 95:e2086-e2094. [PMID: 32847955 PMCID: PMC7713748 DOI: 10.1212/wnl.0000000000010707] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/29/2020] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE To examine in vivo amyloid burden in relation to APOEε4 genotype in middle-aged Hispanics. We hypothesize higher amyloid levels among APOE ε4 carriers vs APOE ε4 noncarriers. METHODS This is a cross-sectional study in a community-based sample of 249 middle-aged Hispanics in New York City who underwent a 3T brain MRI and PET with the amyloid radioligand 18F-florbetaben. APOE genotype was the primary exposure. The primary outcome was amyloid positivity. The secondary outcome was subthreshold amyloid levels examined as a continuous variable. RESULTS APOE ε4 carriers (n = 85) had a higher frequency (15.3%) of amyloid positivity compared to APOE ε4 noncarriers (n = 164, 1.8%). In the subthreshold group of amyloid-negative participants (n = 233), APOE ε4 carriers (n = 72) had a 0.02 (95% confidence interval [CI] 0.01-0.04) higher global brain amyloid standardized uptake value ratio (SUVR) compared to APOE ε4 noncarriers (n = 161). Compared to participants with the ε3/ε3 genotype, participants with ε4/ε4 had the highest frequency of amyloid positivity (28.6%), followed by those with ε3/ε4 (11%). Among amyloid-negative participants (n = 233), compared to participants with ε3/ε3 (n = 134), those with ε4/ε4 (n = 5) had a 0.12 (95% CI 0.07-0.17) higher global brain amyloid SUVR, and those with ε3/ε4 had a 0.02 higher SUVR (95% CI 0.003-0.04). Results were similar when a median split was used for elevated amyloid, when continuous amyloid SUVR was analyzed in all participants, and in nonparametric Mann-Whitney comparisons. CONCLUSION Middle-aged Hispanic APOE ε4 carriers have higher in vivo brain amyloid burden compared with noncarriers, as reported in non-Hispanics.
Collapse
Affiliation(s)
- Priya Palta
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY.
| | - Brady Rippon
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Christiane Reitz
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Hengda He
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Greysi Sherwood
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Fernando Ceballos
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Jeanne Teresi
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Qolamreza Razlighi
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Herman Moreno
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - Adam M Brickman
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| | - José A Luchsinger
- From the Department of Medicine (P.P., B.R., G.S., F.C., J.A.L.), Vagelos College of Physicians and Surgeons, Department of Epidemiology (P.P., C.R., J.A.L.), Joseph P. Mailman School of Public Health, Department of Neurology (C.R., H.H., Q.R., A.M.B.), College of Physicians and Surgeons, Taub Institute for Research on Alzheimer's Disease and the Aging Brain (C.R., Q.R., A.M.B.), and Gertrude H. Sergievsky Center (C.R., Q.R., A.M.B.), Columbia University Irving Medical Center, New York; Research Division (J.T.), Hebrew Home in Riverdale, Bronx; Columbia University Stroud Center at New York State Psychiatric Center (J.T.); Department of Biomedical Engineering (Q.R.), Columbia University, New York; and Department of Neurology (H.M.), SUNY Downstate Medical Center, Brooklyn, NY
| |
Collapse
|
56
|
de Frutos-Lucas J, Cuesta P, Ramírez-Toraño F, Nebreda A, Cuadrado-Soto E, Peral-Suárez Á, Lopez-Sanz D, Bruña R, Marcos-de Pedro S, Delgado-Losada ML, López-Sobaler AM, Concepción Rodríguez-Rojo I, Barabash A, Serrano Rodriguez JM, Laws SM, Dolado AM, López-Higes R, Brown BM, Maestú F. Age and APOE genotype affect the relationship between objectively measured physical activity and power in the alpha band, a marker of brain disease. Alzheimers Res Ther 2020; 12:113. [PMID: 32962736 PMCID: PMC7507658 DOI: 10.1186/s13195-020-00681-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Electrophysiological studies show that reductions in power within the alpha band are associated with the Alzheimer's disease (AD) continuum. Physical activity (PA) is a protective factor that has proved to reduce AD risk and pathological brain burden. Previous research has confirmed that exercise increases power in the alpha range. However, little is known regarding whether other non-modifiable risk factors for AD, such as increased age or APOE ε4 carriage, alter the association between PA and power in the alpha band. METHODS The relationship between PA and alpha band power was examined in a sample of 113 healthy adults using magnetoencephalography. Additionally, we explored whether ε4 carriage and age modulate this association. The correlations between alpha power and gray matter volumes and cognition were also investigated. RESULTS We detected a parieto-occipital cluster in which PA positively correlated with alpha power. The association between PA and alpha power remained following stratification of the cohort by genotype. Younger and older adults were investigated separately, and only younger adults exhibited a positive relationship between PA and alpha power. Interestingly, when four groups were created based on age (younger-older adult) and APOE (E3/E3-E3/E4), only younger E3/E3 (least predicted risk) and older E3/E4 (greatest predicted risk) had associations between greater alpha power and higher PA. Among older E3/E4, greater alpha power in these regions was associated with improved memory and preserved brain structure. CONCLUSION PA could protect against the slowing of brain activity that characterizes the AD continuum, where it is of benefit for all individuals, especially E3/E4 older adults.
Collapse
Affiliation(s)
- Jaisalmer de Frutos-Lucas
- Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
- Biological and Health Psychology Department, School of Psychology, Universidad Autonoma de Madrid, 28049, Madrid, Spain.
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain.
| | - Pablo Cuesta
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
| | - Federico Ramírez-Toraño
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
| | - Alberto Nebreda
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
| | - Esther Cuadrado-Soto
- Departamento de Nutricion y Ciencia de los Alimentos, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
- IMDEA-Food, CEI UAM + CSIC, Madrid, 28049, Spain
| | - África Peral-Suárez
- Departamento de Nutricion y Ciencia de los Alimentos, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - David Lopez-Sanz
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Department of Psychobiology and Methodology in Behavioral Sciences, Universidad Complutense de Madrid (UCM), Pozuelo de Alarcón, 28223, Spain
| | - Ricardo Bruña
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Silvia Marcos-de Pedro
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Departamento de Especialidades Medicas y Salud Pública, Universidad Rey Juan Carlos, 28922, Alcorcon, Spain
| | - María Luisa Delgado-Losada
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
| | - Ana María López-Sobaler
- Departamento de Nutricion y Ciencia de los Alimentos, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Inmaculada Concepción Rodríguez-Rojo
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28040, Madrid, Spain
- Physiotherapy and Nursing Faculty, University of Castilla-La Mancha, Toledo, 45004, Spain
| | - Ana Barabash
- Endocrinology and Nutrition Department, Hospital Clinico San Carlos and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
- Facultad de Psicología, Centro Universitario Villanueva, 28034, Madrid, Spain
| | - Juan Manuel Serrano Rodriguez
- Biological and Health Psychology Department, School of Psychology, Universidad Autonoma de Madrid, 28049, Madrid, Spain
| | - Simon M Laws
- Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Alberto Marcos Dolado
- Neurology Department, Hospital Clinico San Carlos and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Ramón López-Higes
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
| | - Belinda M Brown
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Parque Científico y Tecnológico de la UPM, Crta. M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
- Experimental Psychology Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcon, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| |
Collapse
|
57
|
Gonneaud J, Bedetti C, Pichet Binette A, Benzinger TLS, Morris JC, Bateman RJ, Poirier J, Breitner JCS, Villeneuve S. Association of education with Aβ burden in preclinical familial and sporadic Alzheimer disease. Neurology 2020; 95:e1554-e1564. [PMID: 32759192 PMCID: PMC7713743 DOI: 10.1212/wnl.0000000000010314] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 03/23/2020] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To determine whether years of education and the ε4 risk allele at APOE influence β-amyloid (Aβ) pathology similarly in asymptomatic individuals with a family history of sporadic Alzheimer disease (AD) and presymptomatic autosomal dominant AD mutation carriers. METHODS We analyzed cross-sectional data from 106 asymptomatic individuals with a parental history of sporadic AD (PREVENT-AD cohort; age 67.28 ± 4.72 years) and 117 presymptomatic autosomal dominant AD mutation carriers (DIAN cohort; age 35.04 ± 9.43 years). All participants underwent structural MRI and Aβ-PET imaging. In each cohort we investigated the influence of years of education, APOE ε4 status, and their interaction on Aβ-PET. RESULTS Asymptomatic individuals with a parental history of sporadic AD showed increased Aβ burden associated with APOE ε4 carriage and lower level of education, but no interaction between these. Presymptomatic mutation carriers of autosomal dominant AD showed no relation between APOE ε4 and Aβ burden, but increasing level of education was associated with reduced Aβ burden. The association between educational attainment and Aβ burden was similar in the 2 cohorts. CONCLUSIONS While the APOE ε4 allele confers increased tendency toward Aβ accumulation in sporadic AD only, protective environmental factors, like increased education, may promote brain resistance against Aβ pathology in both sporadic and autosomal dominant AD.
Collapse
Affiliation(s)
- Julie Gonneaud
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO.
| | - Christophe Bedetti
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - Alexa Pichet Binette
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - Tammie L S Benzinger
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - John C Morris
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - Randall J Bateman
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - Judes Poirier
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - John C S Breitner
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO
| | - Sylvia Villeneuve
- From the Department of Psychiatry (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), McGill University; Douglas Mental Health University Institute (J.G., C.B., A.P.B., J.P., J.C.S.B., S.V.), StoP-AD Centre, Montreal, Canada; Knight Alzheimer's Disease Research Center (T.L.S.B., J.C.M., R.J.B.); and Washington University School of Medicine (T.L.S.B., J.C.M., R.J.B.), St. Louis, MO.
| |
Collapse
|
58
|
McGurran H, Glenn JM, Madero EN, Bott NT. Prevention and Treatment of Alzheimer's Disease: Biological Mechanisms of Exercise. J Alzheimers Dis 2020; 69:311-338. [PMID: 31104021 DOI: 10.3233/jad-180958] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. With an aging population and no disease modifying treatments available, AD is quickly becoming a global pandemic. A substantial body of research indicates that lifestyle behaviors contribute to the development of AD, and that it may be worthwhile to approach AD like other chronic diseases such as cardiovascular disease, in which prevention is paramount. Exercise is an important lifestyle behavior that may influence the course and pathology of AD, but the biological mechanisms underpinning these effects remain unclear. This review focuses on how exercise can modify four possible mechanisms which are involved with the pathology of AD: oxidative stress, inflammation, peripheral organ and metabolic health, and direct interaction with AD pathology. Exercise is just one of many lifestyle behaviors that may assist in preventing AD, but understanding the systemic and neurobiological mechanisms by which exercise affects AD could help guide the development of novel pharmaceutical agents and non-pharmacological personalized lifestyle interventions for at-risk populations.
Collapse
Affiliation(s)
- Hugo McGurran
- Research Master's Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Nicholas T Bott
- Neurotrack Technologies Inc., Redwood City, CA, USA.,Clinical Excellence Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychology, PGSP-Stanford Consortium, Palo Alto University, Palo Alto, CA, USA
| |
Collapse
|
59
|
Arellanes IC, Choe N, Solomon V, He X, Kavin B, Martinez AE, Kono N, Buennagel DP, Hazra N, Kim G, D'Orazio LM, McCleary C, Sagare A, Zlokovic BV, Hodis HN, Mack WJ, Chui HC, Harrington MG, Braskie MN, Schneider LS, Yassine HN. Brain delivery of supplemental docosahexaenoic acid (DHA): A randomized placebo-controlled clinical trial. EBioMedicine 2020; 59:102883. [PMID: 32690472 PMCID: PMC7502665 DOI: 10.1016/j.ebiom.2020.102883] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Past clinical trials of docosahexaenoic Acid (DHA) supplements for the prevention of Alzheimer's disease (AD) dementia have used lower doses and have been largely negative. We hypothesized that larger doses of DHA are needed for adequate brain bioavailability and that APOE4 is associated with reduced delivery of DHA and eicosapentaenoic acid (EPA) to the brain before the onset of cognitive impairment. METHODS 33 individuals were provided with a vitamin B complex (1 mg vitamin B12, 100 mg of vitamin B6 and 800 mcg of folic acid per day) and randomized to 2,152 mg of DHA per day or placebo over 6 months. 26 individuals completed both lumbar punctures and MRIs, and 29 completed cognitive assessments at baseline and 6 months. The primary outcome was the change in CSF DHA. Secondary outcomes included changes in CSF EPA levels, MRI hippocampal volume and entorhinal thickness; exploratory outcomes were measures of cognition. FINDINGS A 28% increase in CSF DHA and 43% increase in CSF EPA were observed in the DHA treatment arm compared to placebo (mean difference for DHA (95% CI): 0.08 µg/mL (0.05, 0.10), p<0.0001; mean difference for EPA: 0.008 µg/mL (0.004, 0.011), p<0.0001). The increase in CSF EPA in non-APOE4 carriers after supplementation was three times greater than APOE4 carriers. The change in brain volumes and cognitive scores did not differ between groups. INTERPRETATION Dementia prevention trials using omega-3 supplementation doses equal or lower to 1 g per day may have reduced brain effects, particularly in APOE4 carriers. TRIAL REGISTRATION NCT02541929. FUNDING HNY was supported by R01AG055770, R01AG054434, R01AG067063 from the National Institute of Aging and NIRG-15-361854 from the Alzheimer's Association, and MGH by the L. K. Whittier Foundation. This work was also supported by P50AG05142 (HCC) from the National Institutes of Health. Funders had no role in study design, data collection, data analysis, interpretation, or writing of the report.
Collapse
Affiliation(s)
| | - Nicholas Choe
- Department of Medicine, Keck School of Medicine USC, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine USC, United States
| | - Xulei He
- Department of Medicine, Keck School of Medicine USC, United States
| | - Brian Kavin
- Department of Medicine, Keck School of Medicine USC, United States
| | | | - Naoko Kono
- Department of Preventive Medicine, Keck School of Medicine USC, United States
| | | | - Nalini Hazra
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, USC, United States
| | - Giselle Kim
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, USC, United States
| | - Lina M D'Orazio
- Department of Neurology, Keck School of Medicine USC, United States
| | - Carol McCleary
- Department of Neurology, Keck School of Medicine USC, United States
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Keck School of Medicine USC, United States
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine USC, United States
| | - Howard N Hodis
- Department of Medicine, Keck School of Medicine USC, United States; Department of Preventive Medicine, Keck School of Medicine USC, United States
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine USC, United States
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine USC, United States
| | - Michael G Harrington
- Huntington Medical Research Institutes, CA, United States; Department of Neurology, Keck School of Medicine USC, United States
| | - Meredith N Braskie
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, USC, United States
| | - Lon S Schneider
- Department of Neurology, Keck School of Medicine USC, United States; Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine USC, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine USC, United States; Department of Neurology, Keck School of Medicine USC, United States.
| |
Collapse
|
60
|
Valenzuela PL, Castillo-García A, Morales JS, de la Villa P, Hampel H, Emanuele E, Lista S, Lucia A. Exercise benefits on Alzheimer's disease: State-of-the-science. Ageing Res Rev 2020; 62:101108. [PMID: 32561386 DOI: 10.1016/j.arr.2020.101108] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/29/2020] [Accepted: 06/06/2020] [Indexed: 01/15/2023]
Abstract
Although there is no unanimity, growing evidence supports the value of regular physical exercise to prevent Alzheimer's disease as well as cognitive decline in affected patients. Together with an introductory summary on epidemiological evidence, the aim of this review is to summarize the current knowledge on the potential biological mechanisms underlying exercise benefits in this condition. Regular physical exercise has proven to be beneficial for traditional cardiovascular risk factors (e.g., reduced vascular flow, diabetes) involved in the pathogenesis of Alzheimer's disease. Exercise also promotes neurogenesis via increases in exercise-induced metabolic factors (e.g., ketone bodies, lactate) and muscle-derived myokines (cathepsin-B, irisin), which in turn stimulate the production of neurotrophins such as brain-derived neurotrophic factor. Finally, regular exercise exerts anti-inflammatory effects and improves the brain redox status, thereby ameliorating the pathophysiological hallmarks of Alzheimer's disease (e.g., amyloid-β deposition). In summary, physical exercise might provide numerous benefits through different pathways that might, in turn, help prevent risk and progression of Alzheimer's disease. More evidence is needed, however, based on human studies.
Collapse
|
61
|
Emerging Therapeutic Promise of Ketogenic Diet to Attenuate Neuropathological Alterations in Alzheimer's Disease. Mol Neurobiol 2020; 57:4961-4977. [PMID: 32820459 DOI: 10.1007/s12035-020-02065-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and chronic neurodegenerative disorder that interferes with memory, thinking, and behavior. The consumption of dietary fat has been considered a vital factor for AD as this disease is related to blood-brain barrier function and cholesterol signaling. The ε4 allele of apolipoprotein E (APOE4) is a primary genetic risk factor that encodes one of many proteins accountable for the transport of cholesterol and it is deemed as the leading cholesterol transport proteins in the brain. In case of AD development, the causative factor is the high level of serum/plasma cholesterol. However, this statement is arguable and, in the meantime, the levels of brain cholesterol in individuals with AD are extremely inconstant and levels of cholesterol in the brain and serum/plasma of AD individuals do not reflect cholesterol as a risk factor. In fact, APOE4 is neither fundamental nor sufficient for the advancement of AD; it just acts as a synergistic and increases the danger of AD. Another noticeable characteristic of AD is area-specific decreases in the metabolism of brain glucose. It has been found that the brain cells cannot efficiently metabolize fats; hence, they totally rely upon glucose as a vitality substrate. Thus, suppression of glucose metabolism can possess an intense effect on brain actions. Hypometabolism is frequently found in AD and has quite recently achieved impressive consideration as a plausible target for interfering in the progression of the disease. One promising approach is to keep up the normal supply of glucose to the brain with ketone bodies from the ketogenic diet signifies a potential therapeutic agent for AD. Therefore, this review represents the role of ketogenic diets to combat AD pathogenesis by considering the influence of APOE.
Collapse
|
62
|
Barha CK, Liu-Ambrose T. Sex differences in exercise efficacy: Is midlife a critical window for promoting healthy cognitive aging? FASEB J 2020; 34:11329-11336. [PMID: 32761860 DOI: 10.1096/fj.202000857r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 01/17/2023]
Abstract
Dementia is one of the most pressing health care issues of this century. As no curative treatment for dementia exists, research efforts are growing to identify effective lifestyle interventions to prevent or delay onset. One such promising strategy that promotes cognitive and brain health is engaging in physical exercise. However, current exercise recommendations are imprecise. To advance the potential of exercise as a preventative and treatment strategy, important questions regarding moderators (ie, biological sex and age) are being addressed in the literature. Biological sex is recognized as an important variable to consider in exercise efficacy on brain health, with females showing greater cognitive gains. This may be related to sex differences in underlying mechanisms. Here, we argue to better understand the sex differences in exercise efficacy, the timing of exercise intervention should also be considered. Specifically, we present the hypothesis that midlife in females is a critical window for the implementation of exercise as an early intervention to promote brain health and prevent dementia. Further, we speculate that exercise interventions targeting midlife will be of critical importance for the female brain, as females exit this period of the lifespan at greater risk for cognitive impairment. Given the potential sex differences in dementia risk and prevalence, it is imperative to assess potential sex differences in exercise efficacy as an early intervention during midlife.
Collapse
Affiliation(s)
- Cindy K Barha
- Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada.,Aging, Mobility, and Cognitive Neuroscience Lab, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Physical Activity for Precision Health Research Cluster, University of British Columbia, Vancouver, BC, Canada
| | - Teresa Liu-Ambrose
- Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada.,Aging, Mobility, and Cognitive Neuroscience Lab, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Physical Activity for Precision Health Research Cluster, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
63
|
Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener 2020; 15:40. [PMID: 32677986 PMCID: PMC7364557 DOI: 10.1186/s13024-020-00391-7] [Citation(s) in RCA: 453] [Impact Index Per Article: 113.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder seen in age-dependent dementia. There is currently no effective treatment for AD, which may be attributed in part to lack of a clear underlying mechanism. Studies within the last few decades provide growing evidence for a central role of amyloid β (Aβ) and tau, as well as glial contributions to various molecular and cellular pathways in AD pathogenesis. Herein, we review recent progress with respect to Aβ- and tau-associated mechanisms, and discuss glial dysfunction in AD with emphasis on neuronal and glial receptors that mediate Aβ-induced toxicity. We also discuss other critical factors that may affect AD pathogenesis, including genetics, aging, variables related to environment, lifestyle habits, and describe the potential role of apolipoprotein E (APOE), viral and bacterial infection, sleep, and microbiota. Although we have gained much towards understanding various aspects underlying this devastating neurodegenerative disorder, greater commitment towards research in molecular mechanism, diagnostics and treatment will be needed in future AD research.
Collapse
Affiliation(s)
- Tiantian Guo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Denghong Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yuzhe Zeng
- Department of Orthopaedics, Orthopaedic Center of People's Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou, China
| | - Timothy Y Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Yingjun Zhao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
64
|
Liccardo D, Marzano F, Carraturo F, Guida M, Femminella GD, Bencivenga L, Agrimi J, Addonizio A, Melino I, Valletta A, Rengo C, Ferrara N, Rengo G, Cannavo A. Potential Bidirectional Relationship Between Periodontitis and Alzheimer's Disease. Front Physiol 2020; 11:683. [PMID: 32719612 PMCID: PMC7348667 DOI: 10.3389/fphys.2020.00683] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia in the elderly population, representing a global public health priority. Despite a large improvement in understanding the pathogenesis of AD, the etiology of this disorder remains still unclear, and no current treatment is able to prevent, slow, or stop its progression. Thus, there is a keen interest in the identification and modification of the risk factors and novel molecular mechanisms associated with the development and progression of AD. In this context, it is worth noting that several findings support the existence of a direct link between neuronal and non-neuronal inflammation/infection and AD progression. Importantly, recent studies are now supporting the existence of a direct relationship between periodontitis, a chronic inflammatory oral disease, and AD. The mechanisms underlying the association remain to be fully elucidated, however, it is generally accepted, although not confirmed, that oral pathogens can penetrate the bloodstream, inducing a low-grade systemic inflammation that negatively affects brain function. Indeed, a recent report demonstrated that oral pathogens and their toxic proteins infect the brain of AD patients. For instance, when AD progresses from the early to the more advanced stages, patients could no longer be able to adequately adhere to proper oral hygiene practices, thus leading to oral dysbiosis that, in turn, fuels infection, such as periodontitis. Therefore, in this review, we will provide an update on the emerging (preclinical and clinical) evidence that supports the relationship existing between periodontitis and AD. More in detail, we will discuss data attesting that periodontitis and AD share common risk factors and a similar hyper-inflammatory phenotype.
Collapse
Affiliation(s)
- Daniela Liccardo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Translational Medicine, Temple University, Philadelphia, PA, United States
| | - Federica Marzano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | | | - Marco Guida
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Jacopo Agrimi
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Armida Addonizio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Imma Melino
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessandra Valletta
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Carlo Rengo
- Department of Prosthodontics and Dental Materials, School of Dental Medicine, University of Siena, Siena, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Istituti Clinici Scientifici ICS Maugeri - S.p.A.-Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Scientifico di Telese Terme, Telese, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Istituti Clinici Scientifici ICS Maugeri - S.p.A.-Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Scientifico di Telese Terme, Telese, Italy
| | - Alessandro Cannavo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
65
|
Stojanovic M, Jin Y, Fagan AM, Benzinger TL, Hassenstab J, Cruchaga C, Morris JC, Head D. Physical Exercise and Longitudinal Trajectories in Alzheimer Disease Biomarkers and Cognitive Functioning. Alzheimer Dis Assoc Disord 2020; 34:212-219. [PMID: 32520736 PMCID: PMC7483844 DOI: 10.1097/wad.0000000000000385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Associations of physical exercise with Alzheimer disease (AD) biomarkers and cognitive functioning have been observed cross-sectionally. However, the effects of exercise on longitudinal change in AD biomarkers have not been thoroughly investigated. The current study examined whether individuals with higher baseline exercise exhibited less longitudinal change in AD biomarkers and cognitive functioning, and whether APOE and/or brain-derived neurotrophic factor (BDNF) genotypes moderated the effects of exercise on longitudinal changes. METHODS Clinically normal individuals completed a questionnaire on physical exercise over the prior 10-year period at baseline. Ninety-five individuals had serial cerebrospinal fluid samples collected to examine Aβ42, ptau181 and total tau; 181 individuals underwent multiple assessments of amyloid positron emission tomography imaging with Pittsburgh Compound-B; 327 individuals underwent multiple cognitive assessments, including measures of episodic memory, executive functions, verbal fluency, and processing speed. RESULTS Greater exercise was associated with less steep decline in processing speed. Baseline exercise did not robustly impact longitudinal change for any other outcomes. Neither APOE nor BDNF genotype robustly moderated the effect of exercise on trajectories of AD biomarkers or cognitive decline. INTERPRETATION Results suggest that self-reported physical exercise may be limited as a moderator of changes in AD biomarkers.
Collapse
Affiliation(s)
| | | | - Anne M Fagan
- Knight Alzheimer Disease Research Center
- Hope Center for Neurological Disorders
- Department of Neurology
| | | | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri 63130, United States of America
| | - John C Morris
- Knight Alzheimer Disease Research Center
- Department of Neurology
| | - Denise Head
- Department of Psychological and Brain Sciences
- Knight Alzheimer Disease Research Center
- Department of Radiology
| |
Collapse
|
66
|
ApoE Genotype-Dependent Response to Antioxidant and Exercise Interventions on Brain Function. Antioxidants (Basel) 2020; 9:antiox9060553. [PMID: 32630431 PMCID: PMC7346214 DOI: 10.3390/antiox9060553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
This study determined whether antioxidant supplementation is a viable complement to exercise regimens in improving cognitive and motor performance in a mouse model of Alzheimer’s disease risk. Starting at 12 months of age, separate groups of male and female mice expressing human Apolipoprotein E3 (GFAP-ApoE3) or E4 (GFAP-ApoE4) were fed either a control diet or a diet supplemented with vitamins E and C. The mice were further separated into a sedentary group or a group that followed a daily exercise regimen. After 8 weeks on the treatments, the mice were administered a battery of functional tests including tests to measure reflex and motor, cognitive, and affective function while remaining on their treatment. Subsequently, plasma inflammatory markers and catalase activity in brain regions were measured. Overall, the GFAP-ApoE4 mice exhibited poorer motor function and spatial learning and memory. The treatments improved balance, learning, and cognitive flexibility in the GFAP-ApoE3 mice and overall the GFAP-ApoE4 mice were not responsive. The addition of antioxidants to supplement a training regimen only provided further benefits to the active avoidance task, and there was no antagonistic interaction between the two interventions. These outcomes are indicative that there is a window of opportunity for treatment and that genotype plays an important role in response to interventions.
Collapse
|
67
|
Effects of Physical Activity on Brain Energy Biomarkers in Alzheimer's Diseases. Diseases 2020; 8:diseases8020018. [PMID: 32521816 PMCID: PMC7349237 DOI: 10.3390/diseases8020018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 11/29/2022] Open
Abstract
The prevalence of dementia has substantially increased worldwide. Currently, there is no cure for dementia or Alzheimer’s disease (AD), and care for affected patients is financially and psychologically costly. Of late, more attention has been given to preventive interventions—in particular, physical activity/exercise. In this review, examine the risk factors associated with AD and the effects physical activity may play in the prevention of the degenerative process of this disease, loss of memory and cognitive performance in the elderly. To date, research has shown that physical activity, especially aerobic exercise, has a protective effect on cognitive function and memory in the elderly and Alzheimer’s patients. In comparison with aerobic exercise, several strength training studies have also shown positive effects, and the rare studies that compare the two different modalities show no difference.
Collapse
|
68
|
Kimura N, Aso Y, Yabuuchi K, Ishibashi M, Hori D, Sasaki Y, Nakamichi A, Uesugi S, Jikumaru M, Sumi K, Eguchi A, Obara H, Kakuma T, Matsubara E. Association of Modifiable Lifestyle Factors With Cortical Amyloid Burden and Cerebral Glucose Metabolism in Older Adults With Mild Cognitive Impairment. JAMA Netw Open 2020; 3:e205719. [PMID: 32515796 PMCID: PMC7284299 DOI: 10.1001/jamanetworkopen.2020.5719] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPORTANCE Understanding the association of lifestyle factors with mild cognitive impairment enables the development of evidence-based interventions for delaying cognitive impairment. OBJECTIVE To explore whether objectively measured lifestyle factors, such as physical activity, conversation, and sleep, are associated with cortical amyloid burden and cerebral glucose metabolism in older adults with mild cognitive impairment. DESIGN, SETTING, AND PARTICIPANTS This cohort study included 855 community-dwelling adults in Usuki, Oita Prefecture, Japan, aged 65 years or older. Data were collected from August 2015 to December 2017. Participants were reviewed to examine risk and protective lifestyle factors for dementia. Data analysis was conducted in June 2019. EXPOSURES Wearable sensors, carbon-11 labeled Pittsburgh compound B positron emission tomography images, and fluorine-18 fluorodeoxyglucose positron emission tomography images. MAIN OUTCOMES AND MEASURES Wearable sensor data, such as walking steps, conversation time, and sleep, were collected from August 2015 to October 2017, and positron emission tomography images were collected from October 2015 to December 2017. A multiple regression model and change-point regression model were used to examine the association of lifestyle factors with mean amyloid or fluorodeoxyglucose uptake, assessed on the basis of a standardized uptake value ratio of the frontal lobes, temporoparietal lobes, and posterior cingulate gyrus with the cerebellar cortex as the reference region. The bootstrap method was used to obtain nonparametric 95% CIs on the associations of lifestyle factors with cognitive decline. RESULTS Of the 855 adults in the study, 118 (13.8%) were diagnosed with mild cognitive impairment, with a mean (SD) age of 75.7 (5.8) years and 66 (55.9%) women. Total sleep time was inversely associated with fluorodeoxyglucose uptake after adjusting for covariates (β = -0.287; 95% CI, -0.452 to -0.121, P < .001). Change-point regression showed an inverse association between total sleep time and mean amyloid uptake when sleep duration was longer than 325 minutes (B = -0.0018; 95% CI, -0.0031 to -0.0007). CONCLUSIONS AND RELEVANCE To our knowledge, this is the first study to demonstrate that total sleep time was associated with brain function in older adults with mild cognitive impairment. Sleep duration is a potentially modifiable risk factor for dementia at the mild cognitive impairment stage.
Collapse
Affiliation(s)
- Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yasuhiro Aso
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenichi Yabuuchi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masato Ishibashi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Daiji Hori
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yuuki Sasaki
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Atsuhito Nakamichi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Souhei Uesugi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mika Jikumaru
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kaori Sumi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | - Atsuko Eguchi
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| | | | | | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
69
|
Tsolaki AC, Tsolaki M, Pandria N, Lazarou E, Gkatzima O, Zilidou V, Karagianni M, Iakovidou-Kritsi Z, Kimiskidis VK, Bamidis PD. Web-Based Intervention Effects on Mild Cognitive Impairment Based on Apolipoprotein E Genotype: Quasi-Experimental Study. J Med Internet Res 2020; 22:e14617. [PMID: 32379048 PMCID: PMC7243129 DOI: 10.2196/14617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/19/2019] [Accepted: 12/15/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Apolipoprotein E (APOE) ε4 allele is a major genetic risk factor for Alzheimer disease and mild cognitive impairment (MCI). Computer-based training programs can improve cognitive performance in elderly populations. However, the effects of computer-based interventions on MCI APOE ε4 carriers have never been studied before. OBJECTIVE The effects of different web-based interventions and the APOE isoform-specific differences in training outcomes are investigated. METHODS Using a quasi-experimental study design, 202 participants with MCI aged 60 years and older took part in three different intervention programs (physical and cognitive [Long-Lasting Memories, or LLM], cognitive [Active Control, or AC], or physical intervention [Physical Training Control, or PTC]) via an innovative information and communication technologies exergaming platform. Participants in each interventional group were subdivided into APOE ε4 carriers and non-APOE ε4 carriers. All participants underwent an extensive neuropsychological evaluation before and after the training, blood tests, and brain imaging. RESULTS All interventions resulted in multiple statistically significant cognitive benefits after the intervention. Verbal learning (California Verbal Learning Test: immediate recall test score-LLM: P=.04; AC: P<.001), working memory (digit span forward and backward test scores-AC: P=.03; PTC: P=.02 and P=.006, respectively), and long-term memory (California Verbal Learning Test: delayed recall test score-LLM: P=.02; AC: P=.002; and PTC: P=.02) were improved. There was no statistically significant difference among the intervention effects. APOE ε4 presence moderates intervention effects as the LLM intervention improved only their task-switching processing speed (Trail Making Test, Part B: P=.03) and the PTC intervention improved only the working memory (digit span backward: P=.03). No significant performance alteration was noted for the APOE ε4+ cognitive AC training group. CONCLUSIONS None of the applied interventions could be identified as the optimal one; it is suggested, however, that combined cognitive and physical training and physical training via exergaming may be more effective for the high-risk MCI ΑPOE ε4+ subgroup.
Collapse
Affiliation(s)
- Anthoula C Tsolaki
- Medical Physics Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Neurology, Agios Pavlos General Hospital, Thessaloniki, Greece
| | - Magda Tsolaki
- 1st Department of Neurology, American Hellenic Educational Progressive Association Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Niki Pandria
- Medical Physics Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eftychia Lazarou
- 1st Department of Neurology, American Hellenic Educational Progressive Association Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Olymbia Gkatzima
- Panhellenic Institute of Neurodegenerative Diseases, Thessaloniki, Greece
| | - Vasiliki Zilidou
- Medical Physics Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Karagianni
- Medical Physics Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Zafiroula Iakovidou-Kritsi
- Laboratory of Medical Biology-Genetics Department, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vasilios K Kimiskidis
- Laboratory of Clinical Neurophysiology, American Hellenic Educational Progressive Association Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Panagiotis D Bamidis
- Medical Physics Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
70
|
Iso-Markku P, Waller K, Hautasaari P, Kaprio J, Kujala UM, Tarkka IM. Twin studies on the association of physical activity with cognitive and cerebral outcomes. Neurosci Biobehav Rev 2020; 114:1-11. [PMID: 32325068 DOI: 10.1016/j.neubiorev.2020.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 02/25/2020] [Accepted: 04/09/2020] [Indexed: 11/24/2022]
Abstract
Regular physical activity (PA) offers positive effects on the human body. However, the effects of PA on cognition and in the brain are less clear. In this paper, we narratively review the relationship of PA with cognition and dementia, first from general perspective and then through genetically informed studies on the topic. Then we move on to imaging studies on exercise and brain anatomy first by presenting an overall picture of the topic and then discussing brain imaging studies addressing PA and brain structure in twins in more detailed way. Regarding PA and cognition or dementia, genetically informed studies are uncommon, even though the relationship between PA and cognitive ageing has been extensively studied. It is challenging to find twin pairs discordant for PA and dementia. Concerning brain imaging studies, among PA discordant young adult twin pairs, the more active co-twins showed larger gray matter volumes in striatal, prefrontal, and hippocampal regions and in electrophysiological studies automatic deviance-detection processes differed in brain regions involved with sensorimotor, visual and memory functions.
Collapse
Affiliation(s)
- Paula Iso-Markku
- Department of Clinical Physiology and Nuclear Medicine, HUS Medical Imaging Center, Helsinki 42, University Central Hospital and University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Katja Waller
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Pekka Hautasaari
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Urho M Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Ina M Tarkka
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| |
Collapse
|
71
|
Mueller KD, Norton D, Koscik RL, Morris MC, Jonaitis EM, Clark LR, Fields T, Allison S, Berman S, Kraning S, Zuelsdorff M, Okonkwo O, Chin N, Carlsson CM, Bendlin BB, Hermann BP, Johnson SC. Self-reported health behaviors and longitudinal cognitive performance in late middle age: Results from the Wisconsin Registry for Alzheimer's Prevention. PLoS One 2020; 15:e0221985. [PMID: 32324741 PMCID: PMC7179879 DOI: 10.1371/journal.pone.0221985] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/16/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Studies have suggested associations between self-reported engagement in health behaviors and reduced risk of cognitive decline. Most studies explore these relationships using one health behavior, often cross-sectionally or with dementia as the outcome. In this study, we explored whether several individual self-reported health behaviors were associated with cognitive decline when considered simultaneously, using data from the Wisconsin Registry for Alzheimer's Prevention (WRAP), an Alzheimer's disease risk-enriched cohort who were non-demented and in late midlife at baseline. METHOD We analyzed longitudinal cognitive data from 828 participants in WRAP, with a mean age at baseline cognitive assessment of 57 (range = 36-78, sd = 6.8) and an average of 6.3 years (standard deviation = 1.9, range = 2-10) of follow-up. The primary outcome was a multi-domain cognitive composite, and secondary outcomes were immediate/delayed memory and executive function composites. Predictors of interest were self-reported measures of physical activity, cognitive activity, adherence to a Mediterranean-style diet (MIND), and interactions with each other and age. We conducted linear mixed effects analyses within an Information-theoretic (IT) model averaging (MA) approach on a set of models including covariates and combinations of these 2- and 3-way interactions. The IT approach was selected due to the large number of interactions of interest and to avoid pitfalls of traditional model selection approaches. RESULTS Model-averaged results identified no significant self-reported health behavior*age interactions in relationship to the primary composite outcome. In secondary outcomes, higher MIND diet scores associated with slower decline in executive function. Men showed faster decline than women on delayed memory, independent of health behaviors. There were no other significant interactions among any other health behaviors and cognitive trajectories. CONCLUSIONS When multiple covariates and health behaviors were considered simultaneously, there were limited weak associations with cognitive decline in this age range. These results may be explained alone or in combination by three alternative explanations: 1) the range of cognitive decline is in middle age is too small to observe relationships with health behaviors, 2) the putative associations of these health behaviors on cognition may not be robust in this age range, or 3) the self-reported measures of the health behaviors may not be optimal for predicting cognitive decline. More study may be needed that incorporates sensitive measures of health behaviors, AD biomarker profiles, and/or other disease comorbidities.
Collapse
Affiliation(s)
- Kimberly D. Mueller
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Department of Communication Sciences and Disorders, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Derek Norton
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Rebecca L. Koscik
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Martha C. Morris
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, United States of America
| | - Erin M. Jonaitis
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Lindsay R. Clark
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Department of Neurology, University of Wisconsin–Madison, Madison, WI, United States of America
- Geriatric Research and Education Center, William S. Middleton Memorial Veteran’s Hospital, Madison, WI, United States of America
| | - Taylor Fields
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Samantha Allison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Geriatric Research and Education Center, William S. Middleton Memorial Veteran’s Hospital, Madison, WI, United States of America
| | - Sara Berman
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Sarah Kraning
- Department of Communication Sciences and Disorders, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Megan Zuelsdorff
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Ozioma Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Nathaniel Chin
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Geriatric Research and Education Center, William S. Middleton Memorial Veteran’s Hospital, Madison, WI, United States of America
| | - Barbara B. Bendlin
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Bruce P. Hermann
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Department of Neurology, University of Wisconsin–Madison, Madison, WI, United States of America
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States of America
- Geriatric Research and Education Center, William S. Middleton Memorial Veteran’s Hospital, Madison, WI, United States of America
| |
Collapse
|
72
|
Jeon SY, Byun MS, Yi D, Lee JH, Ko K, Sohn BK, Lee JY, Ryu SH, Lee DW, Shin SA, Kim YK, Kang KM, Sohn CH, Lee DY. Midlife Lifestyle Activities Moderate APOE ε4 Effect on in vivo Alzheimer's Disease Pathologies. Front Aging Neurosci 2020; 12:42. [PMID: 32256335 PMCID: PMC7093017 DOI: 10.3389/fnagi.2020.00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/07/2020] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate whether the midlife cognitive activity and physical activity moderate the relationship between apolipoprotein Eε4 (APOE4) and in vivo Alzheimer's disease (AD) pathologies. In total, 287 non-demented older adults (mean age 72 years) from the Korean Brain Aging Study for the Early diagnosis and prediction of Alzheimer's disease cohort were included. Participants underwent a comprehensive clinical assessment including the evaluation for midlife CA and physical activity, [11C]-Pittsburgh-Compound-B-positron emission tomography (PET), [18F]-fluorodeoxyglucose PET, structural magnetic resonance imaging (MRI), and APOE genotyping. We used linear regression and regression-based mediated-moderation models for statistical analyses. Neither midlife cognitive activity nor physical activity moderated the effect of APOE4 on β-amyloid (Aβ) retention itself. Midlife cognitive activity significantly moderated the effect of APOE4 on hippocampal volume [B (SE) = - 627.580 (252.327), t = -2.488, p = 0.014]: APOE4 carriers had smaller hippocampal volume than non-carriers at relatively high cognitive activity state (p = 0.004), but not at relatively low cognitive activity condition (p = 0.937). Midlife physical activity significantly moderated the effect of Aβ retention, which was closely related to APOE4, on AD-signature region cerebral glucose metabolism [AD-CM; B (SE) = 0.004 (0.002), t = 2.030, p = 0.043]: higher Aβ accumulation was associated with lower AD-CM in relatively low physical activity condition (p < 0.001), whereas no such association was observed in relatively high physical activity state (p = 0.791). The findings suggest that high midlife cognitive activity may accelerate hippocampal atrophy induced by APOE4, whereas high midlife physical activity may delay AD-related cerebral hypometabolism by weakening the influence of APOE4-associated Aβ retention.
Collapse
Affiliation(s)
- So Yeon Jeon
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, South Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jun-Ho Lee
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Kang Ko
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Bo Kyung Sohn
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Seung-Ho Ryu
- Department of Psychiatry, School of Medicine, Konkuk University Medical Center, Konkuk University, Seoul, South Korea
| | - Dong Woo Lee
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Seoung A Shin
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.,Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
73
|
Williams T, Borchelt DR, Chakrabarty P. Therapeutic approaches targeting Apolipoprotein E function in Alzheimer's disease. Mol Neurodegener 2020; 15:8. [PMID: 32005122 PMCID: PMC6995170 DOI: 10.1186/s13024-020-0358-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
One of the primary genetic risk factors for Alzheimer’s disease (AD) is the presence of the Ɛ4 allele of apolipoprotein E (APOE). APOE is a polymorphic lipoprotein that is a major cholesterol carrier in the brain. It is also involved in various cellular functions such as neuronal signaling, neuroinflammation and glucose metabolism. Humans predominantly possess three different allelic variants of APOE, termed E2, E3, and E4, with the E3 allele being the most common. The presence of the E4 allele is associated with increased risk of AD whereas E2 reduces the risk. To understand the molecular mechanisms that underlie APOE-related genetic risk, considerable effort has been devoted towards developing cellular and animal models. Data from these models indicate that APOE4 exacerbates amyloid β plaque burden in a dose-dependent manner. and may also enhance tau pathogenesis in an isoform-dependent manner. Other studies have suggested APOE4 increases the risk of AD by mechanisms that are distinct from modulation of Aβ or tau pathology. Further, whether plasma APOE, by influencing systemic metabolic pathways, can also possibly alter CNS function indirectly is not complete;y understood. Collectively, the available studies suggest that APOE may impact multiple signaling pathways and thus investigators have sought therapeutics that would disrupt pathological functions of APOE while preserving or enhancing beneficial functions. This review will highlight some of the therapeutic strategies that are currently being pursued to target APOE4 towards preventing or treating AD and we will discuss additional strategies that holds promise for the future.
Collapse
Affiliation(s)
- Tosha Williams
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA. .,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA. .,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
74
|
Palta P, Heiss G, Sharrett AR, Gabriel KP, Walker K, Evenson KR, Knopman D, Mosley TH, Wong DF, Gottesman RF. Mid- and Late-Life Leisure-Time Physical Activity and Global Brain Amyloid Burden: The Atherosclerosis Risk in Communities (ARIC)-PET Study. J Alzheimers Dis 2020; 76:139-147. [PMID: 32444546 PMCID: PMC8011955 DOI: 10.3233/jad-200152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Physical activity (PA) may slow the development of dementia by reducing the accumulation of amyloid. OBJECTIVE We tested the hypothesis that higher levels of leisure-time PA in mid- or late-life were associated with lower brain amyloid burden in late-life among 326 non-demented participants from the Atherosclerosis Risk in Communities Study of brain florbetapir positron emission tomography (ARIC-PET) ancillary. METHODS Self-reported PA was quantified using a past-year recall, interviewer-administered questionnaire in mid-life (1987-1989, aged 45-64 years) and late-life (2011-2013, aged 67-89 years). Continuous PA estimates were classified as 1) any leisure-time PA participation (yes/no); 2) meeting the 2018 United States' PA guidelines (yes/no); and 3) per 1 standard deviation (SD) higher metabolic equivalent of task (MET) minutes per week (MET·min·wk-1). A brain magnetic resonance imaging scan with Florbetapir PET was performed in late-life. Adjusted odds ratios (OR) of elevated amyloid burden, defined as a global cortical standardized uptake value ratio (>1.2), compared to no elevated amyloid burden were estimated according to PA measures. RESULTS Among the 326 participants (mean age: 76 years, 42% male, 41% Black), 52% had elevated brain amyloid burden. Mid-life leisure-time PA did not show a statistically significant lower odds of elevated late-life amyloid burden (OR = 0.71, 95% CI: 0.43-1.18). A 1 SD (970 MET. min. wk-1) higher PA level in mid-life was also not significantly associated withelevated amyloid burden (OR = 0.89, 95% CI: 0.69-1.15). Similar estimates were observed for meeting versus not meeting PA guidelines in both mid- and late-life. CONCLUSION Self-reported higher mid- and late-life leisure-time PA were not significantly associated with lower amyloid burden. Data show a trend of an association, which is, however, imprecise, suggesting replication in larger studies.
Collapse
Affiliation(s)
- Priya Palta
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - A. Richey Sharrett
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kelley Pettee Gabriel
- Department of Epidemiology, School of Public Health, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Keenan Walker
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly R. Evenson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Thomas H. Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Dean F. Wong
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Washington University in St. Louis, School of Medicine, Mallinckrodt Institute of Radiology, St. Louis, MO, USA
| | - Rebecca F. Gottesman
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
75
|
Piccarducci R, Daniele S, Fusi J, Chico L, Baldacci F, Siciliano G, Bonuccelli U, Franzoni F, Martini C. Impact of ApoE Polymorphism and Physical Activity on Plasma Antioxidant Capability and Erythrocyte Membranes. Antioxidants (Basel) 2019; 8:E538. [PMID: 31717561 PMCID: PMC6912376 DOI: 10.3390/antiox8110538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
The allele epsilon 4 (ε4) of apolipoprotein E (ApoE) is the strongest genetic risk factor for Alzheimer's disease (AD). ApoE protein plays a pivotal role in the synthesis and metabolism of amyloid beta (Aβ), the major component of the extracellular plaques that constitute AD pathological hallmarks. Regular exercise is an important preventive/therapeutic tool in aging and AD. Nevertheless, the impact of physical exercise on the well-being of erythrocytes, a good model of oxidative stress and neurodegenerative processes, remains to be investigated, particularly depending on ApoE polymorphism. Herein, we evaluate the oxidative status, Aβ levels, and the membrane's composition of erythrocytes in a cohort of human subjects. In our hands, the plasma antioxidant capability (AOC), erythrocytes membrane fluidity, and the amount of phosphatidylcholine (PC) were demonstrated to be significantly decreased in the ApoE ε4 genotype and non-active subjects. In contrast, erythrocyte Aβ content and lipid peroxidation increased in ε4 carriers. Regular physical exercise was associated with an increased plasma AOC and membrane fluidity, as well as to a reduced amount of erythrocytes Aβ. Altogether, these data highlight the influence of the ApoE genotype on erythrocytes' well-being and confirm the positive impact of regular physical exercise.
Collapse
Affiliation(s)
- Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Lucia Chico
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy; (J.F.); (L.C.); (F.B.); (G.S.); (U.B.)
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (R.P.); (S.D.)
| |
Collapse
|
76
|
Berkowitz CL, Mosconi L, Rahman A, Scheyer O, Hristov H, Isaacson RS. Clinical Application of APOE in Alzheimer's Prevention: A Precision Medicine Approach. JPAD-JOURNAL OF PREVENTION OF ALZHEIMERS DISEASE 2019; 5:245-252. [PMID: 30298183 DOI: 10.14283/jpad.2018.35] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Population-attributable risk models estimate that up to one-third of Alzheimer's disease (AD) cases may be preventable through risk factor modification. The field of AD prevention has largely focused on addressing these factors through universal risk reduction strategies for the general population. However, targeting these strategies in a clinical precision medicine fashion, including the use of genetic risk factors, allows for potentially greater impact on AD risk reduction. Apolipoprotein E (APOE), and specifically the APOE ε4 variant, is one of the most well-established genetic influencers on late-onset AD risk. In this review, we evaluate the impact of APOE ε4 carrier status on AD prevention interventions, including lifestyle, nutrigenomic, pharmacogenomic, AD comorbidities, and other biological and behavioral considerations. Using a clinical precision medicine strategy that incorporates APOE ε4 carrier status may provide a highly targeted and distinct approach to AD prevention with greater potential for success.
Collapse
Affiliation(s)
- C L Berkowitz
- Richard S. Isaacson, MD, Department of Neurology, Weill Cornell Medicine and NewYork-Presbyterian, 428 East 72nd St, Suite 500, Room 407, New York, NY, 10021; Tel: (212) 746-3645,
| | | | | | | | | | | |
Collapse
|
77
|
Hansson O, Svensson M, Gustavsson AM, Andersson E, Yang Y, Nägga K, Hållmarker U, James S, Deierborg T. Midlife physical activity is associated with lower incidence of vascular dementia but not Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2019; 11:87. [PMID: 31630687 PMCID: PMC6802179 DOI: 10.1186/s13195-019-0538-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/10/2019] [Indexed: 12/05/2022]
Abstract
Background Physical activity might reduce the risk of developing dementia. However, it is still unclear whether the protective effect differs depending on the subtype of dementia. We aimed to investigate if midlife physical activity affects the development of vascular dementia (VaD) and Alzheimer’s disease (AD) differently in two large study populations with different designs. Methods Using a prospective observational design, we studied whether long-distance skiers of the Swedish Vasaloppet (n = 197,685) exhibited reduced incidence of VaD or AD compared to matched individuals from the general population (n = 197,684) during 21 years of follow-up (median 10, interquartile range (IQR) 5–15 years). Next, we studied the association between self-reported physical activity, stated twice 5 years apart, and incident VaD and AD in 20,639 participants in the Swedish population-based Malmo Diet and Cancer Study during 18 years of follow-up (median 15, IQR 14–17 years). Finally, we used a mouse model of AD and studied brain levels of amyloid-β, synaptic proteins, and cognitive function following 6 months of voluntary wheel running. Results Vasaloppet skiers (median age 36.0 years [IQR 29.0–46.0], 38% women) had lower incidence of all-cause dementia (adjusted hazard ratio (HR) 0.63, 95% CI 0.52–0.75) and VaD (adjusted HR 0.49, 95% CI 0.33–0.73), but not AD, compared to non-skiers. Further, faster skiers exhibited a reduced incidence of VaD (adjusted HR 0.38, 95% CI 0.16–0.95), but not AD or all-cause dementia compared to slower skiers. In the Malmo Diet and Cancer Study (median age 57.5 years [IQR 51.0–63.8], 60% women), higher physical activity was associated with reduced incidence of VaD (adjusted HR 0.65, 95% CI 0.49-0.87), but not AD nor all-cause dementia. These findings were also independent of APOE-ε4 genotype. In AD mice, voluntary running did not improve memory, amyloid-β, or synaptic proteins. Conclusions Our results indicate that physical activity in midlife is associated with lower incidence of VaD. Using three different study designs, we found no significant association between physical activity and subsequent development of AD.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Martina Svensson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
| | - Anna-Märta Gustavsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Emelie Andersson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Ulf Hållmarker
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Stefan James
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden.
| |
Collapse
|
78
|
The Role of Physical Fitness in Cognitive-Related Biomarkers in Persons at Genetic Risk of Familial Alzheimer's Disease. J Clin Med 2019; 8:jcm8101639. [PMID: 31591322 PMCID: PMC6832576 DOI: 10.3390/jcm8101639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/28/2022] Open
Abstract
Introduction: Nondemented people with a family history of Alzheimer’s disease (ADFH) and the ApoE-4 allele have been demonstrated to show a trend for a higher probability of cognitive decline and aberrant levels of cognitive-related biomarkers. However, the potential interactive effects on physical fitness have not been investigated. Purpose: The primary purpose of this study was to determine whether ADFH individuals with the ApoE-4 genotype show deviant brain event-related neural oscillatory performance and cognitively-related molecular indices. A secondary purpose was to examine the interactive effects on physical fitness. Methods: Blood samples were provided from 110 individuals with ADFH to assess molecular biomarkers and the ApoE genotype for the purpose of dividing them into an ApoE-4 group (n = 16) and a non-ApoE-4 group (n = 16) in order for them to complete a visuospatial working memory task while simultaneously recording electroencephalographic signals. They also performed a senior functional physical fitness (SFPF) test. Results: While performing the cognitive task, the ApoE-4 relative to non-ApoE-4 group showed worse accuracy rates (ARs) and brain neural oscillatory performance. There were no significant between-group differences with regard to any molecular biomarkers (e.g., IL-1β, IL-6, IL-8, BDNF, Aβ1-40, Aβ1-42). VO2max was significantly correlated with the neuropsychological performance (i.e., ARs and RTs) in the 2-item and 4-item conditions in the ApoE-4 group and across the two groups. However, the electroencephalogram (EEG) oscillations during visuospatial working memory processing in the two conditions were not correlated with any SFPF scores or cardiorespiratory tests in the two groups. Conclusions: ADFH individuals with the ApoE-4 genotype only showed deviant neuropsychological (e.g., ARs) and neural oscillatory performance when performing the cognitive task with a higher visuospatial working memory load. Cardiorespiratory fitness potentially played an important role in neuropsychological impairment in this group.
Collapse
|
79
|
Villeneuve S. Lifespan Cognitive Reserve—A Secret to Coping With Neurodegenerative Pathology. JAMA Neurol 2019; 76:1145-1146. [DOI: 10.1001/jamaneurol.2019.2899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Sylvia Villeneuve
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health Research Institute, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
80
|
Abstract
Technologies for imaging the pathophysiology of Alzheimer disease (AD) now permit studies of the relationships between the two major proteins deposited in this disease - amyloid-β (Aβ) and tau - and their effects on measures of neurodegeneration and cognition in humans. Deposition of Aβ in the medial parietal cortex appears to be the first stage in the development of AD, although tau aggregates in the medial temporal lobe (MTL) precede Aβ deposition in cognitively healthy older people. Whether aggregation of tau in the MTL is the first stage in AD or a fairly benign phenomenon that may be transformed and spread in the presence of Aβ is a major unresolved question. Despite a strong link between Aβ and tau, the relationship between Aβ and neurodegeneration is weak; rather, it is tau that is associated with brain atrophy and hypometabolism, which, in turn, are related to cognition. Although there is support for an interaction between Aβ and tau resulting in neurodegeneration that leads to dementia, the unknown nature of this interaction, the strikingly different patterns of brain Aβ and tau deposition and the appearance of neurodegeneration in the absence of Aβ and tau are challenges to this model that ultimately must be explained.
Collapse
|
81
|
Erickson KI, Grove GA, Burns JM, Hillman CH, Kramer AF, McAuley E, Vidoni ED, Becker JT, Butters MA, Gray K, Huang H, Jakicic JM, Kamboh MI, Kang C, Klunk WE, Lee P, Marsland AL, Mettenburg J, Rogers RJ, Stillman CM, Sutton BP, Szabo-Reed A, Verstynen TD, Watt JC, Weinstein AM, Wollam ME. Investigating Gains in Neurocognition in an Intervention Trial of Exercise (IGNITE): Protocol. Contemp Clin Trials 2019; 85:105832. [PMID: 31465859 PMCID: PMC6815730 DOI: 10.1016/j.cct.2019.105832] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Despite the ubiquity of normal age-related cognitive decline there is an absence of effective approaches for improving neurocognitive health. Fortunately, moderate intensity exercise is a promising method for improving brain and cognitive health in late life, but its effectiveness remains a matter of skepticism and debate because of the absence of large, comprehensive, Phase III clinical trials. Here we describe the protocol for such a randomized clinical trial called IGNITE (Investigating Gains in Neurocognition in an Intervention Trial of Exercise), a study capable of more definitively addressing whether exercise influences cognitive and brain health in cognitively normal older adults. We are conducting a 12-month, multi-site, randomized dose-response exercise trial in 639 cognitively normal adults between 65 and 80 years of age. Participants are randomized to (1) a moderate intensity aerobic exercise condition of 150 min/week (N = 213), (2) a moderate intensity aerobic exercise condition at 225 min/week (N = 213), or (3) a light intensity stretching-and-toning control condition for 150 min/week (N = 213). Participants are engaging in 3 days/week of supervised exercise and two more days per week of unsupervised exercise for 12 months. A comprehensive cognitive battery, blood biomarkers and battery of psychosocial questionnaires is assessed at baseline, 6 and 12-months. In addition, brain magnetic resonance imaging, physiological biomarkers, cardiorespiratory fitness, physical function, and positron emission tomography of amyloid deposition are assessed at baseline and at the 12-month follow-up. The results from this trial could transform scientific-based policy and health care recommendations for approaches to improve cognitive function in cognitively normal older adults.
Collapse
Affiliation(s)
- Kirk I Erickson
- Department of Psychology, University of Pittsburgh, USA.; Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Australia.
| | | | - Jeffrey M Burns
- Department of Neurology, University of Kansas Medical Center, USA
| | - Charles H Hillman
- Department of Psychology, Northeastern University, USA; Department of Physical Therapy, Movement, & Rehabilitation Sciences, Northeastern University, USA
| | - Arthur F Kramer
- Department of Psychology, Northeastern University, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, USA
| | - Edward McAuley
- Department of Kinesiology and Community Health, University of Illinois at Urbana Champaign, USA
| | - Eric D Vidoni
- Department of Neurology, University of Kansas Medical Center, USA
| | - James T Becker
- Department of Psychology, University of Pittsburgh, USA.; Department of Psychiatry, University of Pittsburgh, USA; Department of Neurology, University of Pittsburgh, USA
| | | | - Katerina Gray
- Department of Psychology, University of Pittsburgh, USA
| | - Haiqing Huang
- Department of Psychology, University of Pittsburgh, USA
| | - John M Jakicic
- Department of Health and Physical Activity, University of Pittsburgh, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, USA
| | - Chaeryon Kang
- Department of Biostatistics, University of Pittsburgh, USA
| | | | - Phil Lee
- Department of Radiology, University of Kansas Medical Center, USA
| | | | | | - Renee J Rogers
- Department of Health and Physical Activity, University of Pittsburgh, USA
| | | | - Bradley P Sutton
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, USA
| | - Amanda Szabo-Reed
- Department of Internal Medicine, University of Kansas Medical Center, USA
| | | | | | | | | |
Collapse
|
82
|
Dunn AR, O'Connell KMS, Kaczorowski CC. Gene-by-environment interactions in Alzheimer's disease and Parkinson's disease. Neurosci Biobehav Rev 2019; 103:73-80. [PMID: 31207254 PMCID: PMC6700747 DOI: 10.1016/j.neubiorev.2019.06.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
Diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) arise from complex interactions of genetic and environmental factors, with genetic variants regulating individual responses to environmental exposures (i.e. gene-by-environment interactions). Identifying gene-by-environment interactions will be critical to fully understanding disease mechanisms and developing personalized therapeutics, though these interactions are still poorly understood and largely under-studied. Candidate gene approaches have shown that known disease risk variants often regulate response to environmental factors. However, recent improvements in exposome- and genome-wide association and interaction studies in humans and mice are enabling discovery of novel genetic variants and pathways that predict response to a variety of environmental factors. Here, we highlight recent approaches and ongoing developments in human and rodent studies to identify genetic modulators of environmental factors using AD and PD as exemplars. Identifying gene-by-environment interactions in disease will be critical to developing personalized intervention strategies and will pave the way for precision medicine.
Collapse
Affiliation(s)
- Amy R Dunn
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| | | | | |
Collapse
|
83
|
Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol 2019; 15:501-518. [PMID: 31367008 DOI: 10.1038/s41582-019-0228-7] [Citation(s) in RCA: 697] [Impact Index Per Article: 139.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 02/06/2023]
Abstract
Polymorphism in the apolipoprotein E (APOE) gene is a major genetic risk determinant of late-onset Alzheimer disease (AD), with the APOE*ε4 allele conferring an increased risk and the APOE*ε2 allele conferring a decreased risk relative to the common APOE*ε3 allele. Strong evidence from clinical and basic research suggests that a major pathway by which APOE4 increases the risk of AD is by driving earlier and more abundant amyloid pathology in the brains of APOE*ε4 carriers. The number of amyloid-β (Aβ)-dependent and Aβ-independent pathways that are known to be differentially modulated by APOE isoforms is increasing. For example, evidence is accumulating that APOE influences tau pathology, tau-mediated neurodegeneration and microglial responses to AD-related pathologies. In addition, APOE4 is either pathogenic or shows reduced efficiency in multiple brain homeostatic pathways, including lipid transport, synaptic integrity and plasticity, glucose metabolism and cerebrovascular function. Here, we review the recent progress in clinical and basic research into the role of APOE in AD pathogenesis. We also discuss how APOE can be targeted for AD therapy using a precision medicine approach.
Collapse
|
84
|
Ettore E, Bakardjian H, Solé M, Levy Nogueira M, Habert MO, Gabelle A, Dubois B, Robert P, David R. Relationships between objectives sleep parameters and brain amyloid load in subjects at risk for Alzheimer’s disease: the INSIGHT-preAD Study. Sleep 2019; 42:5527427. [DOI: 10.1093/sleep/zsz137] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
Study Objectives
Sleep changes have been associated with increased risks of developing cognitive disturbances and Alzheimer’s disease (AD). A bidirectional relation is underlined between amyloid-beta (Aß) and sleep disruptions. The sleep profile in participants at risk to develop AD is not fully deciphered. We aim to investigate sleep–wake changes with objective sleep measurements in elderly participants without cognitive impairment depending on their brain amyloid status, positive (Aß+) or negative (Aß−) based on standard absorption ratios (SUVr) positron emission tomography-florbetapir imaging.
Methods
Sixty-eight participants without cognitive impairment who have accepted to be involved in the sleep ancillary study from the InveStIGation of Alzheimer’s Predictors in Subjective Memory Complainers (INSIGHT-pre AD) cohort, aiming to record sleep profile based on the analyses of an ambulatory accelerometer-based assessment (seven consecutive 24-hour periods). Neuropsychological tests were performed and sleep parameters have been individualized by actigraph. Participants also underwent a magnetic resonance imaging scan to assess their hippocampal volume. Based on SUVr PET-florbetapir imaging, two groups Aß+ and Aß− were compared.
Results
Participants were divided into two groups: Aß+ (n = 24) and Aß− (n = 44). Except for the SUVr, the two subgroups were comparable. When looking to sleep parameters, increased sleep latency, sleep fragmentation (wake after sleep onset [WASO] score and awakenings) and worst sleep efficiency were associated with cortical brain amyloid load.
Conclusion
Actigraphic sleep parameters were associated with cortical brain amyloid load in participants at risk to develop AD. The detection of sleep abnormalities in those participants may be of interest to propose some preventive strategies.
Collapse
Affiliation(s)
- Eric Ettore
- Université Côte d’Azur, CoBTeK lab, Centre Mémoire de Ressources et de Recherche, Nice University Hospital France
- Department of Psychiatry, Nice University Hospital France
| | - Hovagim Bakardjian
- Institute of Memory and Alzheimer’s Disease, ICM, INSERM, CNRS UMR, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University Paris, Paris, France
| | - Marine Solé
- Institute of Memory and Alzheimer’s Disease, ICM, INSERM, CNRS UMR, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University Paris, Paris, France
| | - Marcel Levy Nogueira
- Institute of Memory and Alzheimer’s Disease, ICM, INSERM, CNRS UMR, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University Paris, Paris, France
| | - Marie-Odile Habert
- Nuclear Medicine Department, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University Paris, Paris, France
| | - Audrey Gabelle
- Memory Research and Resources Center for Alzheimer’s disease, Department of Neurology, Montpellier University Hospital, and Inserm, and University of Montpellier, Montpellier, France
| | - Bruno Dubois
- Institute of Memory and Alzheimer’s Disease, ICM, INSERM, CNRS UMR, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University Paris, Paris, France
| | - Philippe Robert
- Université Côte d’Azur, CoBTeK lab, Centre Mémoire de Ressources et de Recherche, Nice University Hospital France
| | - Renaud David
- Université Côte d’Azur, CoBTeK lab, Centre Mémoire de Ressources et de Recherche, Nice University Hospital France
| |
Collapse
|
85
|
Brown BM, Rainey-Smith SR, Dore V, Peiffer JJ, Burnham SC, Laws SM, Taddei K, Ames D, Masters CL, Rowe CC, Martins RN, Villemagne VL. Self-Reported Physical Activity is Associated with Tau Burden Measured by Positron Emission Tomography. J Alzheimers Dis 2019; 63:1299-1305. [PMID: 29758940 DOI: 10.3233/jad-170998] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Numerous animal studies have reported exercise reduces the accumulation of Alzheimer's disease pathology, including amyloid-β (Aβ) and tau. Furthermore, we previously reported a relationship between higher levels of physical activity (PA) and lower brain Aβ burden in a human population. The recent advent of tau positron emission tomography (PET) tracers enables us to extend our investigations into the evaluation of the relationship between PA and brain tau burden. Utilizing data from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study, we have examined the cross-sectional relationship between habitual PA and PET-quantified tau burden. Forty-three cognitively healthy older adults were categorized into low-moderate PA (LMPA; n = 16) or high PA (HPA; n = 27), based on self-reported PA levels. Tau PET imaging with the AV1451 tracer was conducted on all participants. The LMPA group had significantly higher neocortical tau burden (presented as a z-score; 1.22±1.98), compared to the HPA group (z-score: - 0.28±1.18). The difference between the LMPA and HPA groups was also evident when examining regional tau burden in the temporoparietal cortex and the prefrontal cortex. Our results suggest an association between self-reported PA level and brain tau burden. Future longitudinal and interventional studies utilizing larger samples sizes are vital to further investigate the nature of the relationship between tau and PA.
Collapse
Affiliation(s)
- Belinda M Brown
- School of Psychology and Exercise Science, Murdoch University, Murdoch, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Nedlands, WA, Australia
| | - Stephanie R Rainey-Smith
- Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Nedlands, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Vincent Dore
- eHealth, CSIRO Health and Biosecurity, Herston, QLD, Australia
| | - Jeremiah J Peiffer
- School of Psychology and Exercise Science, Murdoch University, Murdoch, WA, Australia
| | | | - Simon M Laws
- Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Nedlands, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, WA, Australia.,Cooperative Research Centre for Mental Health, http://www.mentalhealthcrc.com
| | - Kevin Taddei
- Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Nedlands, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - David Ames
- Department of Psychiatry, University of Melbourne, VIC, Australia.,National Ageing Research Institute, Parkville, VIC, Australia
| | - Colin L Masters
- Florey Institute for Neurosciences and Mental Health, University of Melbourne, VIC, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Ralph N Martins
- Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Nedlands, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| |
Collapse
|
86
|
Frederiksen KS, Madsen K, Andersen BB, Beyer N, Garde E, Høgh P, Waldemar G, Hasselbalch SG, Law I. Moderate- to high-intensity exercise does not modify cortical β-amyloid in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:208-215. [PMID: 31198839 PMCID: PMC6556817 DOI: 10.1016/j.trci.2019.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Animal models of Alzheimer's disease show that exercise may modify β-amyloid (Aβ) deposition. We examined the effect of a 16-week exercise intervention on cortical Aβ in patients with mild-to-moderate Alzheimer's disease. METHODS Thirty-six patients with Alzheimer's disease were randomized to either one hour of aerobic exercise three times weekly for 16 weeks or usual care. Pre and post intervention, 11Carbon-Pittsburgh compound B positron emission tomography was carried out to assess cortical Aβ, and quantified using standardized uptake value rations (SUVRs). RESULTS The intervention showed no effect on follow-up SUVRs in a covariance analysis with group allocation, baseline intervention SUVR, age, sex, and baseline Mini-Mental State Examination as predictors. Change in SUVRs did not correlate with changes in measures of physical or aerobic fitness. DISCUSSION The present findings do not support an effect of exercise on Aβ. However, the relatively short intervention period may account for a lack of efficacy. Further studies should test earlier and longer interventions.
Collapse
Affiliation(s)
- Kristian S. Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Karine Madsen
- Neurobiology Research Unit, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte B. Andersen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nina Beyer
- Musculoskeletal Rehabilitation Research Unit and Institute of Sports Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Garde
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Public Health and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Peter Høgh
- Zealand University Hospital, Department of Neurology, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen G. Hasselbalch
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
87
|
Lyall DM, Celis-Morales C, Lyall LM, Graham C, Graham N, Mackay DF, Strawbridge RJ, Ward J, Gill JMR, Sattar N, Cavanagh J, Smith DJ, Pell JP. Assessing for interaction between APOE ε4, sex, and lifestyle on cognitive abilities. Neurology 2019; 92:e2691-e2698. [PMID: 31028125 PMCID: PMC6556094 DOI: 10.1212/wnl.0000000000007551] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 02/04/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To test for interactions between APOE ε4 genotype and lifestyle factors on worse cognitive abilities in UK Biobank. METHODS Using UK Biobank cohort data, we tested for interactions between APOE ε4 allele presence, lifestyle factors of alcohol intake, smoking, total physical activity and obesity, and sex, on cognitive tests of reasoning, information processing speed, and executive function (n range = 70,988-324,725 depending on the test). We statistically adjusted for potential confounders of age, sex, deprivation, cardiometabolic conditions, and educational attainment. RESULTS There were significant associations between APOE ε4 and worse cognitive abilities, independent of potential confounders, and between lifestyle risk factors and worse cognitive abilities; however, there were no interactions at multiple correction-adjusted p < 0.05, against our hypotheses. CONCLUSIONS Our results do not provide support for the idea that ε4 genotype increases vulnerability to the negative effects of lifestyle risk factors on cognitive ability, but rather support a primarily outright association between APOE ε4 genotype and worse cognitive ability.
Collapse
Affiliation(s)
- Donald M Lyall
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden.
| | - Carlos Celis-Morales
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Laura M Lyall
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Christopher Graham
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Nicholas Graham
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Daniel F Mackay
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Rona J Strawbridge
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Joey Ward
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Jason M R Gill
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Naveed Sattar
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Jonathan Cavanagh
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Daniel J Smith
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| | - Jill P Pell
- From the Institute of Health & Wellbeing (D.M.L., L.M.L., C.G., N.G., D.F.M., R.J.S., J.W., J.C., D.J.S., J.P.P.) and Institute of Cardiovascular and Medical Sciences (C.C.-M., J.M.R.G., N.S.), University of Glasgow, Scotland, UK; and Department of Medicine Solna (R.J.S.), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
88
|
Nilsson MI, Tarnopolsky MA. Mitochondria and Aging-The Role of Exercise as a Countermeasure. BIOLOGY 2019; 8:biology8020040. [PMID: 31083586 PMCID: PMC6627948 DOI: 10.3390/biology8020040] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/15/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022]
Abstract
Mitochondria orchestrate the life and death of most eukaryotic cells by virtue of their ability to supply adenosine triphosphate from aerobic respiration for growth, development, and maintenance of the ‘physiologic reserve’. Although their double-membrane structure and primary role as ‘powerhouses of the cell’ have essentially remained the same for ~2 billion years, they have evolved to regulate other cell functions that contribute to the aging process, such as reactive oxygen species generation, inflammation, senescence, and apoptosis. Biological aging is characterized by buildup of intracellular debris (e.g., oxidative damage, protein aggregates, and lipofuscin), which fuels a ‘vicious cycle’ of cell/DNA danger response activation (CDR and DDR, respectively), chronic inflammation (‘inflammaging’), and progressive cell deterioration. Therapeutic options that coordinately mitigate age-related declines in mitochondria and organelles involved in quality control, repair, and recycling are therefore highly desirable. Rejuvenation by exercise is a non-pharmacological approach that targets all the major hallmarks of aging and extends both health- and lifespan in modern humans.
Collapse
Affiliation(s)
- Mats I Nilsson
- Department of Pediatrics and Medicine, McMaster University Medical Center, Hamilton, ON L8S 4L8, Canada.
- Exerkine Corporation, McMaster University Medical Center, Hamilton, ON L8N 3Z5, Canada.
| | - Mark A Tarnopolsky
- Department of Pediatrics and Medicine, McMaster University Medical Center, Hamilton, ON L8S 4L8, Canada.
- Exerkine Corporation, McMaster University Medical Center, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
89
|
Voss MW, Soto C, Yoo S, Sodoma M, Vivar C, van Praag H. Exercise and Hippocampal Memory Systems. Trends Cogn Sci 2019; 23:318-333. [PMID: 30777641 PMCID: PMC6422697 DOI: 10.1016/j.tics.2019.01.006] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 01/17/2023]
Abstract
No medications prevent or reverse age-related cognitive decline. Physical activity (PA) enhances memory in rodents, but findings are mixed in human studies. As a result, exercise guidelines specific for brain health are absent. Here, we re-examine results from human studies, and suggest the use of more sensitive tasks to evaluate PA effects on age-related changes in the hippocampus, such as relational memory and mnemonic discrimination. We discuss recent advances from rodent and human studies into the underlying mechanisms at both the central and peripheral levels, including neurotrophins and myokines that could contribute to improved memory. Finally, we suggest guidelines for future research to help expedite well-founded PA recommendations for the public.
Collapse
Affiliation(s)
- Michelle W Voss
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA.
| | - Carmen Soto
- Laboratory of Neurogenesis and Neuroplasticity, Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Seungwoo Yoo
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Matthew Sodoma
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Carmen Vivar
- Laboratory of Neurogenesis and Neuroplasticity, Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Henriette van Praag
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
90
|
Duara R, Loewenstein DA, Lizarraga G, Adjouadi M, Barker WW, Greig-Custo MT, Rosselli M, Penate A, Shea YF, Behar R, Ollarves A, Robayo C, Hanson K, Marsiske M, Burke S, Ertekin-Taner N, Vaillancourt D, De Santi S, Golde T, St D. Effect of age, ethnicity, sex, cognitive status and APOE genotype on amyloid load and the threshold for amyloid positivity. NEUROIMAGE-CLINICAL 2019; 22:101800. [PMID: 30991618 PMCID: PMC6447735 DOI: 10.1016/j.nicl.2019.101800] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/08/2019] [Accepted: 03/26/2019] [Indexed: 11/30/2022]
Abstract
The threshold for amyloid positivity by visual assessment on PET has been validated by comparison to amyloid load measured histopathologically and biochemically at post mortem. As such, it is now feasible to use qualitative visual assessment of amyloid positivity as an in-vivo gold standard to determine those factors which can modify the quantitative threshold for amyloid positivity. We calculated quantitative amyloid load, measured as Standardized Uptake Value Ratios (SUVRs) using [18-F]florbetaben PET scans, for 159 Hispanic and non-Hispanic participants, who had been classified clinically as Cognitively Normal (CN), Mild Cognitive Impairment (MCI) or Dementia (DEM). PET scans were visually rated as amyloid positive (A+) or negative (A-), and these judgments were used as the gold standard with which to determine (using ROC analyses) the SUVR threshold for amyloid positivity considering factors such as age, ethnicity (Hispanic versus non-Hispanic), gender, cognitive status, and apolipoprotein E ε4 carrier status. Visually rated scans were A+ for 11% of CN, 39.0% of MCI and 70% of DEM participants. The optimal SUVR threshold for A+ among all participants was 1.42 (sensitivity = 94%; specificity = 92.5%), but this quantitative threshold was higher among E4 carriers (SUVR = 1.52) than non-carriers (SUVR = 1.31). While mean SUVRs did not differ between Hispanic and non-Hispanic participants;, a statistically significant interaction term indicated that the effect of E4 carrier status on amyloid load was greater among non-Hispanics than Hispanics. Visual assessment, as the gold standard for A+, facilitates determination of the effects of various factors on quantitative thresholds for amyloid positivity. A continuous relationship was found between amyloid load and global cognitive scores, suggesting that any calculated threshold for the whole group, or a subgroup, is artefactual and that the lowest calculated threshold may be optimal for the purposes of early diagnosis and intervention. Demographic factors did not affect the threshold for amyloid positivity. Cognitive status did not affect this threshold for amyloid positivity. APOE4 carriers had a higher threshold for amyloid positivity than non-carriers. Among APOE4 carriers, non-Hispanics had higher amyloid load than non- Hispanics. There was a continuous relationship between amyloid load and cognitive status.
Collapse
Affiliation(s)
- R Duara
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA; University of Florida College of Medicine, Gainesville, FL, USA.
| | - D A Loewenstein
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA; Miller School of Medicine, University of Miami, Miami, FL, USA
| | - G Lizarraga
- Florida ADRC, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - M Adjouadi
- Florida ADRC, USA; College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - W W Barker
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - M T Greig-Custo
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - M Rosselli
- Florida ADRC, USA; Florida Atlantic University, USA
| | - A Penate
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - Y F Shea
- Mount Sinai Medical Center, Miami Beach, USA; Department of Medicine, University of Hong Kong, Hong Kong
| | - R Behar
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - A Ollarves
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - C Robayo
- Florida ADRC, USA; Mount Sinai Medical Center, Miami Beach, USA
| | - K Hanson
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | - M Marsiske
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA; University of Florida, College of Public Health and Health Professions, USA
| | - S Burke
- Florida ADRC, USA; Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - N Ertekin-Taner
- Mayo Clinic Florida, Department of Neurology, Jacksonville, FL, USA; Mayo Clinic Florida, Department of Neuroscience, Jacksonville, FL, USA
| | - D Vaillancourt
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | | | - T Golde
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| | - DeKosky St
- Florida ADRC, USA; University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
91
|
Thibeau S, McFall GP, Camicioli R, Dixon RA. Physical Activity and Mobility Differentially Predict Nondemented Executive Function Trajectories: Do Sex and APOE Moderate These Associations? Gerontology 2019; 65:640-648. [PMID: 30909272 DOI: 10.1159/000496442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/22/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In nondemented aging, higher levels of everyday physical activity (EPA) and mobility performance are associated with better executive function (EF) trajectories. However, these associations may be moderated by both sex and Alzheimer's disease (AD) genetic risk. OBJECTIVES In a longitudinal study, we investigate sex differences in (a) EPA and mobility effects on EF performance (level) and change (slope) and (b) AD genetic risk moderation of these associations. METHODS The longitudinal design included nondemented adults (n = 532, mean age = 70.4 years, range 53-95) from the Victoria Longitudinal Study. Using structural equation analyses on an EF latent variable, we tested (a) sex moderation and (b) interactive effects of sex and APOE on observed EPA-EF and mobility-EF performance and change relationships. RESULTS First, we observed independent sex effects for the EPA-EF and mobility-EF predictions. Whereas EPA had a significant effect on EF performance and change only for females, mobility had a significant effect for both sexes. Notably, males with lower mobility levels experienced steeper EF decline than females with lower mobility levels. Second, we observed significant sex × APOE interaction effects. The combination of lower genetic risk and higher EPA benefitted females but not males. In contrast, lower genetic risk and higher mobility benefited both sexes, although male APOE no-risk carriers with lower mobility levels had EF decline patterns that were similar to APOE risk carriers. CONCLUSIONS Longitudinal analyses across a broad band of aging show that sex moderates the effects of both EPA and mobility on EF performance and change. Notably, this moderation occurs differentially across the AD genetic risk status. These results point to a precision health approach to observational and interventional research in which effects of physical activity and mobility on EF trajectories and dementia are examined in the personalized and interactive context of sex and AD risk.
Collapse
Affiliation(s)
- Sherilyn Thibeau
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - G Peggy McFall
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, Canada
| | - Roger A Dixon
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada, .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada,
| |
Collapse
|
92
|
James BD, Bennett DA. Causes and Patterns of Dementia: An Update in the Era of Redefining Alzheimer's Disease. Annu Rev Public Health 2019; 40:65-84. [PMID: 30642228 DOI: 10.1146/annurev-publhealth-040218-043758] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The burden of dementia continues to increase as the population ages, with no disease-modifying treatments available. However, dementia risk appears to be decreasing, and progress has been made in understanding its multifactorial etiology. The 2018 National Institute on Aging-Alzheimer's Association (NIA-AA) research framework for Alzheimer's disease (AD) defines AD as a biological process measured by brain pathology or biomarkers, spanning the cognitive spectrum from normality to dementia. This framework facilitates interventions in the asymptomatic space and accommodates knowledge that many additional pathologies (e.g., cerebrovascular) contribute to the Alzheimer's dementia syndrome. The framework has implications for how we think about risk factors for "AD": Many commonly accepted risk factors are not related to AD pathology and would no longer be considered risk factors for AD. They may instead be related to other pathologies or resilience to pathology. This review updates what is known about causes, risk factors, and changing patterns of dementia, addressing whether they are related to AD pathology/biomarkers, other pathologies, or resilience.
Collapse
Affiliation(s)
- Bryan D James
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612, USA; .,Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612, USA; .,Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| |
Collapse
|
93
|
Karssemeijer EGA, Aaronson JA, Bossers WJR, Donders R, Olde Rikkert MGM, Kessels RPC. The quest for synergy between physical exercise and cognitive stimulation via exergaming in people with dementia: a randomized controlled trial. ALZHEIMERS RESEARCH & THERAPY 2019; 11:3. [PMID: 30611286 PMCID: PMC6320611 DOI: 10.1186/s13195-018-0454-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
Abstract
Background Exercise is often proposed as a non-pharmacological intervention to delay cognitive decline in people with dementia, but evidence remains inconclusive. Previous studies suggest that combining physical exercise with cognitive stimulation may be more successful in this respect. Exergaming is a promising intervention in which physical exercise is combined with cognitively challenging tasks in a single session. The aim of this study was to investigate the effect of exergame training and aerobic training on cognitive functioning in older adults with dementia. Methods A three-armed randomized controlled trial (RCT) compared exergame training, aerobic training and an active control intervention consisting of relaxation and flexibility exercises. Individuals with dementia were randomized and individually trained three times a week during 12 weeks. Cognitive functioning was measured at baseline, after the 12-week intervention period and at 24-week follow-up by neuropsychological assessment. The domains of executive function, episodic memory, working memory and psychomotor speed were evaluated. Test scores were converted into standardized z-scores that were averaged per domain. Between-group differences were analysed with analysis of covariance. Results Data from 115 people with dementia (mean (SD) age = 79.2 (6.9) years; mean (SD) MMSE score = 22.9 (3.4)) were analysed. There was a significant improvement in psychomotor speed in the aerobic and exergame groups compared to the active control group (mean difference domain score (95% CI) aerobic versus control 0.370 (0.103–0.637), p = 0.007; exergame versus control 0.326 (0.081–0.571), p = 0.009). The effect size was moderate (partial η2 = 0.102). No significant differences between the intervention and control groups were found for executive functioning, episodic memory and working memory. Conclusions To our knowledge, this is the first RCT evaluating the effects of exergame training and aerobic training on cognitive functioning in people with dementia. We found that both exergame training and aerobic training improve psychomotor speed, compared to an active control group. This finding may be clinically relevant as psychomotor speed is an important predictor for functional decline. No effects were found on executive function, episodic memory and working memory. Trial registration Netherlands Trial Register, NTR5581. Registered on 7 October 2015. Electronic supplementary material The online version of this article (10.1186/s13195-018-0454-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esther G A Karssemeijer
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Geriatric Medicine, Nijmegen, the Netherlands.,Radboud University Medical Center, Radboudumc Alzheimer Center, Nijmegen, the Netherlands
| | - Justine A Aaronson
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Psychology, Nijmegen, the Netherlands
| | - Willem J R Bossers
- BeweegStrateeg, Groningen, the Netherlands.,Center for Human Movement Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rogier Donders
- Radboud University Medical Center, Department for Health Evidence, Nijmegen, the Netherlands
| | - Marcel G M Olde Rikkert
- Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Geriatric Medicine, Nijmegen, the Netherlands.,Radboud University Medical Center, Radboudumc Alzheimer Center, Nijmegen, the Netherlands
| | - Roy P C Kessels
- Radboud University Medical Center, Radboudumc Alzheimer Center, Nijmegen, the Netherlands. .,Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Department of Medical Psychology, Nijmegen, the Netherlands. .,Center for Cognition, Donders Institute for Brain Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
94
|
Law LL, Sprecher KE, Dougherty RJ, Edwards DF, Koscik RL, Gallagher CL, Carlsson CM, Zetterberg H, Blennow K, Asthana S, Sager MA, Hermann BP, Johnson SC, Cook DB, Bendlin BB, Okonkwo OC. Cardiorespiratory Fitness Modifies Influence of Sleep Problems on Cerebrospinal Fluid Biomarkers in an At-Risk Cohort. J Alzheimers Dis 2019; 69:111-121. [PMID: 30958346 PMCID: PMC6675618 DOI: 10.3233/jad-180291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Previous studies indicate that cardiorespiratory fitness (CRF) and sleep are each favorably associated with Alzheimer's disease (AD) pathophysiology, including reduced amyloid-β (Aβ) and tau pathology. However, few studies have examined CRF and sleep in the same analysis. OBJECTIVE To examine the relationship between sleep and core AD cerebrospinal fluid (CSF) biomarkers among at-risk healthy late-middle-aged adults and determine whether CRF modifies this association. METHODS Seventy-four adults (age = 64.38±5.48, 68.9% female) from the Wisconsin Registry for Alzheimer's Prevention participated. Sleep was evaluated using the Medical Outcomes Study Sleep Scale, specifically the Sleep Problems Index I (SPI), which incorporates domains of sleep disturbance, somnolence, sleep adequacy, and shortness of breath. Higher scores indicate greater sleep problems. To assess CRF, participants underwent a graded exercise test. CSF was collected via lumbar puncture, from which Aβ42, total-tau (t-tau), and phosphorylated-tau (p-tau) were immunoassayed. Regression analyses examined the association between SPI and CSF biomarkers, and the interaction between SPI and CRF on these same biomarkers, adjusting for relevant covariates. RESULTS Higher SPI scores were associated with greater p-tau (p = 0.027) and higher t-tau/Aβ42 (p = 0.021) and p-tau/Aβ42 (p = 0.009) ratios. Analyses revealed significant SPI*CRF interactions for t-tau (p = 0.016), p-tau (p = 0.008), and p-tau/Aβ42 (p = 0.041); with a trend for t-tau/Aβ42 (p = 0.061). Specifically, the relationship between poorer sleep and these biomarkers was significant among less fit individuals, but not among those who were more fit. CONCLUSION In a late-middle-aged at-risk cohort, CRF attenuated the association between poor sleep and levels of select CSF biomarkers. This suggests fitness may play an important role in preventing AD by protecting against pathology, even in impaired sleep.
Collapse
Affiliation(s)
- Lena L. Law
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kate E. Sprecher
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705 USA
- Wisconsin Center for Sleep Medicine and Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719 USA
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53705 USA
| | - Ryan J. Dougherty
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI 53792 USA
| | - Dorothy F. Edwards
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI 53792 USA
| | - Rebecca L. Koscik
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Catherine L. Gallagher
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, WI 53705 USA
| | - Cynthia M. Carlsson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Sanjay Asthana
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
| | - Mark A. Sager
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Bruce P. Hermann
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, WI 53705 USA
| | - Sterling C. Johnson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Dane B. Cook
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI 53792 USA
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Barbara B. Bendlin
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ozioma C. Okonkwo
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| |
Collapse
|
95
|
Abstract
Alzheimer's disease (AD) dementia refers to a particular onset and course of cognitive and functional decline associated with age together with a particular neuropathology. It was first described by Alois Alzheimer in 1906 about a patient whom he first encountered in 1901. Modern clinical diagnostic criteria have been developed, and criteria have also been proposed to recognize preclinical (or presymptomatic) stages of the disease with the use of biomarkers. The primary neuropathology was described by Alzheimer, and in the mid-1980s subsequently evolved into a more specific neuropathologic definition that recognizes the comorbid neuropathologies that frequently contribute to clinical dementia. Alzheimer's disease is now the most common form of neurodegenerative dementia in the United States with a disproportionate disease burden in minority populations. Deficits in the ability to encode and store new memories characterizes the initial stages of the disease. Subsequent progressive changes in cognition and behavior accompany the later stages. Changes in amyloid precursor protein (APP) cleavage and production of the APP fragment beta-amyloid (Aβ) along with hyperphosphorylated tau protein aggregation coalesce to cause reduction in synaptic strength, synaptic loss, and neurodegeneration. Metabolic, vascular, and inflammatory changes, as well as comorbid pathologies are key components of the disease process. Symptomatic treatment offers a modest, clinically measurable effect in cognition, but disease-modifying therapies are desperately needed.
Collapse
Affiliation(s)
- Jose A Soria Lopez
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States
| | - Hector M González
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States
| | - Gabriel C Léger
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States; Shiley-Marcos Alzheimer's Disease Research Center, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
96
|
Müller S, Preische O, Sohrabi HR, Gräber S, Jucker M, Ringman JM, Martins RN, McDade E, Schofield PR, Ghetti B, Rossor M, Fox NN, Graff-Radford NR, Levin J, Danek A, Vöglein J, Salloway S, Xiong C, Benzinger T, Buckles V, Masters CL, Sperling R, Bateman RJ, Morris JC, Laske C. Relationship between physical activity, cognition, and Alzheimer pathology in autosomal dominant Alzheimer's disease. Alzheimers Dement 2018; 14:1427-1437. [PMID: 30266303 PMCID: PMC6322213 DOI: 10.1016/j.jalz.2018.06.3059] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/15/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Little is known about effects of physical activity (PA) in genetically driven early-onset autosomal dominant Alzheimer's disease (AD). METHODS A total of 372 individuals participating at the Dominantly Inherited Alzheimer Network study were examined to evaluate the cross-sectional relationship of PA with cognitive performance, functional status, cognitive decline, and AD biomarkers in cerebrospinal fluid. Mutation carriers were categorized as high or low exercisers according to WHO recommendations. RESULTS Mutation carriers with high PA showed significantly better cognitive and functional performance and significantly less AD-like pathology in cerebrospinal fluid than individuals with low PA. Mutation carriers with high PA scored 3.4 points better on Mini Mental State Examination at expected symptom onset and fulfilled the diagnosis of very mild dementia 15.1 years later compared with low exercisers. DISCUSSION These results support a beneficial effect of PA on cognition and AD pathology even in individuals with genetically driven autosomal dominant AD.
Collapse
Affiliation(s)
- Stephan Müller
- Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany.
| | - Oliver Preische
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Hamid R Sohrabi
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Susanne Gräber
- Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - John M Ringman
- Memory and Aging Center, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Eric McDade
- University of Pittsburgh School of Medicine, Department of Neurology, Pittsburgh, PA, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| | - Martin Rossor
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Nick N Fox
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Neill R Graff-Radford
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), München, Germany; Department of Neurology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Adrian Danek
- German Center for Neurodegenerative Diseases (DZNE), München, Germany; Department of Neurology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), München, Germany; Department of Neurology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stephen Salloway
- Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Chengjie Xiong
- Division of Biostatistics, The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie Benzinger
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Virginia Buckles
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Reisa Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Randall J Bateman
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Christoph Laske
- Section for Dementia Research, Department of Cellular Neurology, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| |
Collapse
|
97
|
Bos MM, Noordam R, Blauw GJ, Slagboom PE, Rensen PCN, van Heemst D. The ApoE ε4 Isoform: Can the Risk of Diseases be Reduced by Environmental Factors? J Gerontol A Biol Sci Med Sci 2018; 74:99-107. [DOI: 10.1093/gerona/gly226] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Maxime M Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, the Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, the Netherlands
| | - Gerard J Blauw
- Department of Internal Medicine, Section of Gerontology and Geriatrics, the Netherlands
| | - P Eline Slagboom
- Department of Medical Statistics and Bioinformatics, Section of Molecular Epidemiology, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, the Netherlands
| |
Collapse
|
98
|
The effect of physical exercise on cerebral blood flow in Alzheimer's disease. NEUROIMAGE-CLINICAL 2018; 20:650-654. [PMID: 30211001 PMCID: PMC6129739 DOI: 10.1016/j.nicl.2018.09.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/20/2018] [Accepted: 09/02/2018] [Indexed: 12/11/2022]
Abstract
In recent years there has been an increasing focus on the relation between cerebrovascular health, physical exercise and Alzheimer's disease. The aim of the current study was to determine the effect of moderate-to-high-intensity aerobic exercise on cerebral blood flow in patients with mild to moderate Alzheimer's disease. Fifty-one patients were randomized to either usual care or moderate-to-high intensity aerobic exercise for 16 weeks. Exercise had no consistent effect on whole brain or regional cerebral blood flow. Sixteen weeks of exercise are, therefore, not sufficient to produce a consistent increase in cerebral blood flow in a relatively small sample of Alzheimer's patients.
Collapse
|
99
|
de Frutos-Lucas J, López-Sanz D, Zuluaga P, Rodríguez-Rojo IC, Luna R, López ME, Delgado-Losada ML, Marcos A, Barabash A, López-Higes R, Maestú F, Fernández A. Physical activity effects on the individual alpha peak frequency of older adults with and without genetic risk factors for Alzheimer’s Disease: A MEG study. Clin Neurophysiol 2018; 129:1981-1989. [DOI: 10.1016/j.clinph.2018.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/29/2018] [Accepted: 06/25/2018] [Indexed: 11/30/2022]
|
100
|
Vos SJ, Visser PJ. Preclinical Alzheimer’s Disease: Implications for Refinement of the Concept. J Alzheimers Dis 2018; 64:S213-S227. [DOI: 10.3233/jad-179943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Stephanie J.B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, Netherlands
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, Netherlands
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|